Note : Les descriptions sont présentées dans la langue officielle dans laquelle elles ont été soumises.
WO 2020/219963
PCT/US2020/029933
DESCRIPTION
METHODS AND COMPOSITIONS FOR TREATING CHRONIC INFLAMMATORY
INJURY, METAPLASIA, DYSPLASIA AND CANCERS OF EPITHELIAL TISSUES
Priority Claim
The present application claims benefit of priority to U.S. Provisional
Applications Serial
Nos. 62/839,152 and 62/924,978, filed April 26, 2019 and October 23, 2019,
respectively, the entire contents of each application being incorporated
herein by
reference.
Federal Funding Disclosure
The invention was made with government support under Grant No. U24CA228550
awarded by the National Institutes of Health. The government has certain
rights in the
invention.
Background
Metaplasia is the replacement of one differentiated cell type with another
mature
differentiated cell type that is not normally present in a specific tissue.
Typically,
metaplasia is triggered by environmental stimuli, which may act in concert
with the
deleterious effects of microorganisms and inflammation. A hallmark of
metaplasia is
a change in cellular identity. Universally, metaplasia is a precursor to low-
grade
dysplasia, which can culminate in high-grade dysplasia and carcinoma. See
Figure 8.
Typically, the risk of a patient developing cancer increases in a pronounced
manner
as an inflammatory disease or metaplasia progresses to dysplasia.
Figure 9 provides a statistical overview of the risk associated with Barret's
Esophagus
(BE). BE is the result of chronic gastroesophageal reflux disease (GERD) and
represents the end stage of the natural course of this disease. It has been
estimated
that 20% of the population in the United States suffers from gastroesophageal
reflux
and that about 10% of these patients are diagnosed with BE. Commonly, BE is
discovered during endoscopy for the evaluation of GERD symptoms.
It is documented that longstanding exposure of esophageal mucosa to gastric
acidity
results in cellular damage of the stratified squamous epithelium and creates
an
1
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
abnormal environment, which stimulates repair in the form of intestinal
epithelial
metaplasia. The consequence is that the stratified squamous epithelium, which
physiologically lines the esophageal mucosa, is replaced by a pathological,
specialized columnar epithelium which is neither of cardiac nor of stomach
type, but
exhibits features of the intestinal type of epithelium. This pathological type
of
epithelium usually demonstrates DNA alterations that predispose to malignancy.
The
alterations in BE are histologically classified into three categories,
depending on
whether or not they exhibit dysplasia: (1) BE without dysplasia; (2) BE with
low-grade
dysplasia; and (3) BE with high-grade dysplasia (HGD). In BE with HGD,
dysplasia is
confined to the mucosa without crossing the basement membrane. If dysplasia
extends beyond the basement membrane into the lamina propria through the in-
coming lymphatic network, it is defined as intramucosal (superficial)
adenocarcinoma,
whereas if it invades the muscularis mucosa layer it becomes invasive
adenocarcinoma. Thus, BE with HGD is considered a precursor of invasive
adenocarcinoma.
Six to twenty percent of patients with BE and HGD are at greatest risk of
developing
adenocarcinoma within a short period of time, ranging from 17 to 35 month at
follow-
up. Esophagectomy specimens from patients with BE and HGD revealed invasive
adenocarcinoma in 30%40% of cases. A recent meta-analysis demonstrated that
patients with BE and HGD developed esophageal adenocarcinoma with an average
incidence of 6 every 100 patients per year, during the first 1.5 to 7 years of
endoscopic
surveillance. Furthermore, the majority of esophageal adenocarcinoma is
thought to
have evolved from cells that have undergone Barrett's metaplasia.
BE is also classified into two categories according to the extent of
intestinal metaplasia
above the gastroesophageal junction: (1) long segment BE, if the extent of the
intestinal epithelium is greater than 3 cm; and (2) short segment BE, if it is
less than 3
cm. Among patients who undergo endoscopy for symptoms of GERD, the incidence
of long segment BE is 3%-5%, whereas short segment BE occurs in 10%-15%.
Whether long and short segment BE share the same pathogenetic alterations or
the
same predisposition to malignancy still remains unclear; however, both
conditions are
currently treated in the same manner.
A common, and invasive, means for treating certain Barrett's Esophagus
patients is
through endoscopic ablation therapy, such as radiofrequency ablation,
photodynamic
2
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
therapy or cryoablation of esophageal tissue. However, despite a reasonably
high
percentage of patients that reach remission after therapy, many of those
patients
relapse within a few years. For other patients, whether because they are
refractory to
ablative therapy or ineligible due to severe co-morbidities, there are even
fewer
treatment options and those that exist still leave a significant need for more
effective
therapies with better results and/or long durations of remission.
Similar metaplasia-to-dysplasia-to-cancer transitions are observed across a
variety of
other epithelial tissues. Metaplasia tends to occur in tissues constantly
exposed to
environmental agents, which are often injurious in nature. For example, the
pulmonary
system (lungs and trachea) and the gastrointestinal tract are common sites of
metaplasia owing to their contacts with air and food, respectively. In the
ovaries, the
dynamic interaction between ovarian surface epithelium and underlying ovarian
stroma appears to be the origin of epithelial differentiation, metaplasia and
finally
malignant transformation.
There is a substantial unmet medical need not only for treatments that are
effective
for cancers of epithelial tissues, but also treatments directed to metaplasia
and
dysplasia of those tissues.
3
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Summary
One aspect of the present disclosure provides a method for treating a patient
suffering
from chronic inflammatory injury, metaplasia, dysplasia or cancer of an
epithelial
tissue, which method comprises administering to the patient an anti-PESC agent
that
selectively kills or inhibits the proliferation or differentiation of
pathogenic epithelial
stem cells (PESCs) relative to normal epithelial stem cells in the tissue in
which the
PESC is found. Representative epithelial tissues include pulmonary,
genitourinary,
gastrointestinal, pancreatic and hepatic tissues.
Another aspect of the disclosure provides a method of reducing proliferation,
survival,
migration, or colony formation ability of PESCs in a subject in need thereof
comprising
contacting the PESC with a therapeutically effective amount of an anti-PESC
agent
that selectively kills or inhibits the proliferation or differentiation of a
PESC population
relative to normal epithelial stem cells in the tissue in which the PESCs are
found.
Another aspect of the disclosure provides a pharmaceutical preparation for
treating
one or more of chronic inflammatory injury, metaplasia, dysplasia or cancer of
an
epithelial tissue, which preparation comprises an anti-PESC agent that
selectively kills
or inhibits the proliferation or differentiation of PESCs relative to normal
epithelial stem
cells. In certain embodiments, the target tissue for treatment is lung. In
certain
embodiments, the target tissue for treatment is a lung tumor, such as for the
treatment
of non-small cell lung carcinoma (NSCLC) or small cell lung carcinoma (SCLC).
In
certain embodiments, the target tissue for treatment is an ovarian, fallopian
and/or
cervical tissue, such as for the treatment of cervical metaplasia, cervical
cancer,
fallopian cancer and/or ovarian cancer (including taxol and/or cisplatin-
resistant
ovarian cancer).
For example, the present disclosure provides a method for treating a patient
suffering
from one or more of esophagitis (including Eosinophilic esophagitis or EoE),
Barrett's
Esophagus, esophageal dysplasia or esophageal cancer, which method comprises
administering to the patient an anti-PESC agent that selectively kills or
inhibits the
proliferation or differentiation of Barrett's Esophagus stem cells (BESC)
relative to
normal esophageal stem cells. In certain embodiments, the patient presents
with
esophagitis. In certain embodiments, the patient presents with Barrett's
Esophagus.
In certain embodiments, the patient presents with esophageal dysplasia. In
certain
4
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
embodiments, the patient presents with esophageal cancer. In certain
embodiments,
the patient presents with esophageal carcinoma, such as esophageal
adenocarcinoma or esophageal squamous cell carcinoma.
Another aspect of the disclosure provides a method of reducing proliferation,
survival,
migration, or colony formation ability of a BESC in a subject in need thereof
comprising
contacting the BESC with a therapeutically effective amount of an anti-PESC
agent
that selectively kills or inhibits the proliferation or differentiation of
BESC relative to
normal esophageal stem cells.
Another aspect of the disclosure provides a pharmaceutical preparation for
treating
one or more of esophagitis, Barrett's esophagus, esophageal dysplasia or
esophageal
cancer, which preparation comprises an anti-PESC agent that selectively kills
or
inhibits the proliferation or differentiation of BESCs relative to normal
esophageal stem
cells. In certain embodiments, the patient presents with esophagitis. In
certain
embodiments, the patient presents with Barrett's Esophagus. In certain
embodiments,
the patient presents with esophageal dysplasia. In certain embodiments, the
patient
presents with esophageal cancer. In certain embodiments, the patient presents
with
esophageal carcinoma, such as esophageal adenocarcinoma or esophageal
squamous cell carcinoma.
Yet another aspect of the disclosure provides a drug eluting device, such as
for treating
one or more of esophagitis, Barrett's esophagus, esophageal dysplasia or
esophageal
cancer, which device comprises drug release means including an anti-PESC agent
that selectively kills or inhibits the proliferation or differentiation of
PESCs relative to
normal epithelial stem cells, which device when deployed in a patient
positions the
drug release means proximal to the luminal surface of the esophagus and
releases
the agent in an amount sufficient to achieve a therapeutically effective
exposure of the
luminal surface to the agent. In certain embodiments, the patient presents
with
esophagitis. In certain embodiments, the patient presents with Barrett's
Esophagus.
In certain embodiments, the patient presents with esophageal dysplasia. In
certain
embodiments, the patient presents with esophageal cancer. In certain
embodiments,
the patient presents with esophageal carcinoma, such as esophageal
adenocarcinoma or esophageal squamous cell carcinoma. Examples of drug eluting
devices are drug eluting stents, drug eluting collars and drug eluting
ballons.
5
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In other embodiments, there are provided drug eluting devices that can be
implanted
proximal to the diseased portion of the luminal surface of the esophagus, such
as
implanted extraluminally (i.e., submucosally or in or on the circular muscle
or
longitudinal muscle) rather than intraluminally.
In certain embodiments, the anti-PESC agent has an IC50 for selectively
killing PESCs
that is 1/5th or less the IC50 for killing normal epithelial stem cells in the
tissue in which
the PESCs are found, more preferably 1/10th, 1/201h, 1/50t11, 1/1001h,
1/2501h, 1,500111 or
even 1/1000th or less the IC50 for killing normal epithelial stem cells.
In certain embodiments, the anti-PESC agent has an 1050 for selectively
killing BESCs
that is 1/5111 or less the ICso for killing normal esophageal stem cells, more
preferably
1/101h, 1/20th, 1/501h, 1/1 0th 1/250t11, 1/500th or even 1/1000th or less the
IC50 for killing
normal esophageal stem cells.
In certain embodiments, the anti-PESC agent has an 1050 for selectively
inhibiting the
proliferation of PESCs that is 115th or less the IC50 for inhibiting normal
epithelial stem
16 cells in the tissue in which the PESCs are found, more preferably
1/10th, 1/20111, 1/50111
,
1/1001h, 1/250111, 1/500th or even 1/1000th or less the IC50 for inhibiting
the proliferation
of normal epithelial stem cells.
In certain embodiments, the anti-PESC agent has an IC50 for selectively
inhibiting the
proliferation of BESCs that is 1/5Ih or less the IC50 for inhibiting the
proliferation of
normal esophageal stem cells, more preferably 1/10th, 1/2011, 1/50111,
1/100th, 1/250th,
1/500th or even 1/1000th or less the 1050 for inhibiting the proliferation of
normal
esophageal stem cells.
In certain embodiments, the anti-PESC agent has an IC50 for selectively
inhibiting the
differentiation of PESCs that is 115th or less the IC50 for inhibiting the
differentiation of
normal epithelial stem cells, more preferably 1/10th, 1/20th, 1/50th, 1/100th,
1/250th,
1/500111 or even 1/1000th or less the IC50 for inhibiting the differentiation
of normal
epithelial stem cells.
In certain embodiments, the anti-PESC agent has an 1050 for selectively
inhibiting the
differentiation of BESCs that is 115th or less the IC50 for inhibiting the
differentiation of
normal esophageal stem cells, more preferably 1/10th, 1/201h, 1/501h, 1/1001h,
1/250th,
1/500111 or even 1/10001h or less the ICso for inhibiting the differentiation
of normal
esophageal stem cells.
6
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In certain embodiments, the anti-PESC agent has a therapeutic index (TI) for
treating
esophagitis, Barrett's Esophagus, esophageal dysplasia and/or esophageal
cancer of
at least 2, and more preferably has a therapeutic index of at least 5, 10, 20,
50, 100,
250, 500 or 1000 for treating esophagitis, Barrett's Esophagus, esophageal
dysplasia
and/or esophageal cancer.
In certain embodiments, the anti-PESC agent has a therapeutic index (TI) for
treating
ovarian, fallopian and or cervical metaplasia or dysplasia of at least 2, and
more
preferably has a therapeutic index of at least 5, 10, 20, 50, 100, 250, 500 or
1000.
In certain embodiments, the anti-PESC agent has a therapeutic index (TI) for
treating
ovarian cancer (such as taxol and/or cisplatin resistant ovarian cancer) of at
least 2,
and more preferably has a therapeutic index of at least 5, 10, 20, 50, 100,
250, 500 or
1000.
In certain embodiments, the anti-PESC agent has a therapeutic index (TI) for
treating
lung cancer (such NSCLC or SCLC) of at least 2, and more preferably has a
therapeutic index of at least 5, 10, 20, 50, 100, 250, 500 or 1000.
In certain embodiments, the anti-PESC agent has a therapeutic index (TI) for
treating
lung metaplasia or dysplasia of at least 2, and more preferably has a
therapeutic index
of at least 5, 10, 20, 50, 100, 250, 50001 1000.
In certain embodiments, the anti-PESC agent inhibits the proliferation or
differentiation
of PESCs, or kills PESCs, with an IC50 of 10-6M or less, more preferably 10-7M
or less,
10-8 M or less or 10-9M or less.
In certain embodiments, the anti-PESC agent inhibits the proliferation or
differentiation
of BESCs, or kills BESCs, with an ICso of 10-6M or less, more preferably 10-7M
or less,
104M or less or 10-9M or less.
In certain embodiments, the anti-PESC agent is administered during or after
endoscopic ablation therapy, such as radiofrequency ablation, photodynamic
therapy
or cryoablation of esophageal tissue.
In certain embodiments, the anti-PESC agent is administered by topical
application,
such as to esophageal tissue, genitourinary tissue or lung tissue.
In certain embodiments, the anti-PESC agent is administered by submucosal
injection,
such as to esophageal tissue, genitourinary tissue or lung tissue.
7
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In certain embodiments, the anti-PESC agent is formulated for submucosal
injection,
such as to esophageal tissue, genitourinary tissue or lung tissue.
In certain embodiments, the anti-PESC agent is formulated for topical
application,
such as to esophageal tissue, genitourinary tissue or lung tissue.
In certain embodiments, the anti-PESC agent is formulated as part of a
bioadhesive
formulation.
In certain embodiments, the anti-PESC agent is formulated as part of a drug-
eluting
particle, drug eluting matrix or drug-eluting gel.
In certain embodiments, the anti-PESC agent is formulated as part of a
bioerodible
drug-eluting particle, bioerodible drug eluting matrix or bioerodible drug-
eluting gel.
In certain embodiments, the disclosure provides a esophageal topical retentive
formulation for topical application to the luminal surface of the esophagus,
comprising
(i) an anti-PESC agent that selectively kills or inhibits the proliferation or
differentiation
of pathogenic epithelial stem cells relative to normal esophageal stem cells,
(ii) a
bioadhesive, and (iii) optionally, one or more pharmaceutically acceptable
excipients.
For instance, the formulation can have a mucosal surface residence half-life
on
esophageal tissue of at least 30 minutes, more preferably at least 60, 120,
180, 240
or even 300 minutes.
For instance, the formulation can produce at least a minimally effective
concentration
(MEC) of the anti-PESC agent in the esophageal tissue to which it is applied
to which
it is applied for at least 30 minutes, more preferably at least 60, 120, 180,
240 or even
300 minutes.
For instance, the formulation can produce anti-PESC agent concentration in the
esophageal tissue to which it is applied with T1/2 of at least 2 hours, more
preferably
at least 4, 6, 8, 10 or even 12 hours.
In certain embodiments, the formulation produces a systemic concentration of
the anti-
PESC agent which is less than 1/3rd the maximum tolerated does (MTD) for that
agent,
and even more preferably less than 1/5th, 1/10th , 1/20th, 1/50th or even
1/100th the
maximum tolerated does (MTD) for that agent.
In certain embodiments, the topical formulation is a viscous bioadhesive
liquid to coat
the esophagus.
8
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In certain embodiments, the topical formulation comprises anti-PESC eluting
multiparticulates, microparticles, nanoparticles or microdiscs
In further embodiments, there is provided bioadhesive nanoparticle having a
polymeric
surface with an adhesive force equivalent to an adhesive force of between 10
N/m2
and 100,000 N/m2 measured on human mucosal surfaces, which nanoparticle
further
includes at least one anti-PESC agent, the anti-PESC agent dispersed therein
or
thereon, wherein the nanoparticle elutes the anti-PESC agents into the mucous
gel
layer when adhered to mucosa! tissue.
The anti-PESC agent(s) may be selected from, for example, a histone
demethylase
inhibitor, A JmjC demethylase inhibitors, a pan-JmjC demethylase inhibitors, a
receptor tyrosine kinase inhibitor, an EGFR inhibitor, a HER2 inhibitor, a
dual
EGFR/HER2 inhibitor, a proteasome inhibitor, an immunoproteasome inhibitor, a
STAT inhibitor, a STAT3 inhibitor, a FLT3 inhibitor, a GSK3 inhibitor, an
HSP90
inhibitor, an HSP70 inhibitor and a dual HSP90/HSP70 inhibitor, or a
combination
thereof,
In certain embodiments, the bioadhesive nanoparticle further includes at least
one
ESO Regenerative agent dispersed therein or thereon, wherein the nanoparticle
elutes
the both the anti-PESC agent and ESO Regenerative agent into the mucous gel
layer
when adhered to mucosal tissue.
In certain embodiments, the ESO Regenerative agent is pan-inhibitor of ABL
kinase
inhibitor, preferably a BCR-ABL kinase inhibitor Exemplary pan-inhibitor
include
imatinib, nilotinib, dasatinib, bosutinib and ponatinib, and is preferably
ponatinib.
In certain embodiments, the ESO Regenerative agent is a BACE inhibitor, an FAK
inhibitor, a VEGR inhibitor or an Ala inhibitor.
In certain embodiments, the submucosal retentive formulation produces a
systemic
concentration of the ESO Regenerative Agent, such as ponatinib, which is less
than
1/3rd the maximum tolerated does (MTD) for that agent, and even more
preferably less
than 1/51h, 1/10Ih , 1/201h, 1/50th or even 1/100th the maximum tolerated does
(MTD) for
that agent.
In still other embodiments, there is provided a submucosal retentive
formulation
comprising at least one anti-PESC agent and one or more pharmaceutically
acceptable excipients, which formulation is injectable submucosally and forms
a
9
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
submucusal depot releasing an effective amount of the anti-PESC agent into the
surrounding tissue.
In certain embodiments, the submucosal retentive formulation is an injectable
thermogel for submucosal injection, comprising at least one anti-PESC agent
and one
or more pharmaceutically acceptable excipients, wherein the thermogel has a
low-
viscosity fluid at room temperature (and easily injected), and becomes a non-
flowing
gel at body temperature after injection.
To illustrate, the anti-PESC agent(s) formulated in the submucosal retentive
formulations may be selected from, for example, a histone demethylase
inhibitor, A
JrnjC demethylase inhibitors, a pan-JmjC demethylase inhibitors, a receptor
tyrosine
kinase inhibitor, an EGFR inhibitor, a HER2 inhibitor, a dual EGFR/HER2
inhibitor, a
proteasome inhibitor, an immunoproteasome inhibitor, a STAT inhibitor, a STAT3
inhibitor, a FLT3 inhibitor, a GSK3 inhibitor, an HSP90 inhibitor, an HSP70
inhibitor
and a dual HSP90/HSP70 inhibitor, or a combination thereof,
In certain embodiments, the submucosal retentive formulations further includes
at
least one ESO Regenerative agent dispersed therein, wherein the submucosal
retentive formulations release the both the anti-PESC agent and ESO
Regenerative
agent into the tissue surrounding the site of submucosal injection.
In certain embodiments, the ESO Regenerative agent is pan-inhibitor of ABL
kinase
inhibitor, preferably a BCR-ABL kinase inhibitor. Exemplary pan-inhibitor
include
imatinib, nilotinib, dasatinib, bosutinib and ponatinib, and is preferably
ponatinib.
In certain embodiments, the ESO Regenerative agent is a BACE inhibitor, an FAK
inhibitor, a VEGR inhibitor or an AKT inhibitor.
For instance, the submucosal retentive formulation can have a submucosal
residence
half-life in esophageal tissue of at least 30 minutes, more preferably at
least 60, 120,
180, 240 or even 300 minutes.
For instance, the submucosal retentive formulation can produce at least a
minimally
effective concentration (MEC) of the anti-PESC agent in the esophageal tissue
in
which it is injected for at least 30 minutes, more preferably at least 60,
120, 180, 240
or even 300 minutes.
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
For instance, the submucosal retentive formulation can produce anti-PESC agent
concentration in esophageal tissue in which it is injected with T1/2 of at
least 2 hours,
more preferably at least 4, 6, 8, 10 or even 12 hours.
The present disclosure also provides submucosal retentive formulations of ESO
Regenerative Agents. For example the formulation can include (i) a BCR-ABL
kinase
inhibitor, and (ii) one or more pharmaceutically acceptable excipients, which
formulation is injectable submucosally and forms a submucusal depot releasing
an
effective amount of the BCR-ABL kinase inhibitor to the surrounding tissue. In
certain
preferred embodiments, the BCR-ABL kinase inhibitor is ponatinib. In certain
preferred
embodiments, the BCR-ABL kinase inhibitor is a FLT3 inhibitor such as
quizartinib
(AC220), crenolanib (CP-868596), nnidostaurin (PKC-412), lestaurtinib (CEP-
701),
4SC-203, TTT-3002, sorafenib (Bay-43-0006), Ponatinib (AP-24534), sunitinib
(SU-
11248), and/or tandutinib (MLN-0518), or (a) pharmaceutically acceptable
salt(s),
solvate(s), and/or hydrate(s) thereof. Preferably, the FMS-like tyrosine
kinase 3 FLT3)
inhibitor is quizartinib (AC220) or pharmaceutically acceptable salt(s),
solvate(s),
and/or hydrate(s) thereof.
In certain embodiments, the submucosal retentive formulation can have a
submucosal
residence half-life in esophageal tissue of at least 30 minutes, more
preferably at least
60, 120, 180, 240 or even 300 minutes.
In certain embodiments, the submucosal retentive formulation can produce at
least a
minimally effective concentration (MEC) of the ESO Regenerative Agent in the
esophageal tissue in which it is injected for at least 30 minutes, more
preferably at
least 60, 120, 180, 240 or even 300 minutes.
In certain embodimentsõ the submucosal retentive formulation can produce an
ESO
Regenerative Agent concentration in esophageal tissue in which it is injected
with T1/2
of at least 2 hours, more preferably at least 4, 6, 8, 10 or even 12 hours.
In certain embodiments, the submucosal retentive formulation produces a
systemic
concentration of the ESO Regenerative Agent, such as ponatinib, which is less
than
1/31d the maximum tolerated does (MTD) for that agent, and even more
preferably less
than 1/51h, oh,1/1 1/20th, 1/50th or even 1/100th the maximum
tolerated does (MTD) for
that agent.
11
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In each of the above submucosal retentive formulations, the formulation can
form a
flowable and/or viscous gel.
In certain embodiments, the formulation is an injectable thermogel. Thermogels
includes, merely to illustrate, poly(lactic acid-co-glycolic
acid)¨poly(ethylene glycol)-
poly(lactic acid-co-glycolic acid) (PLGA¨PEG¨PLGA) triblock copolymers.
In certain embodiments, the formulation is a hydrogel.
In certain embodiments, the formulation is suitable for endoscopic dissection.
In certain embodiments, the formulation further comprises an anticoagulant.
In certain embodiments, the formulation further comprises comprises one or
more
antitussives, antihistamines, antipyretics, analgesics, anti-infective agents
and/or
chemotherapeutic agents
Another aspect of the present disclosure provides an injectable thermogel for
subnnucosal injection, comprising ponatinib and (optionally) one or more
pharmaceutically acceptable excipients, wherein the thermogel has a low-
viscosity
fluid at room temperature (and easily injected), and becomes a non-flowing gel
at body
temperature after injection.
In certain embodiments, the disclosure provides an esophageal topical
retentive
formulation for topical application to the luminal surface of the esophagus,
comprising
(i) an ESO Regenerative Agent, (ii) a bioadhesive, and (iii) optionally, one
or more
pharmaceutically acceptable excipients. For example the formulation can
include (i)
a BCR-ABL kinase inhibitor, and (ii) one or more pharmaceutically acceptable
excipients, which formulation is injectable submucosally and forms a
submucusal
depot releasing an effective amount of the BCR-ABL kinase inhibitor to the
surrounding tissue. In certain preferred embodiments, the BCR-ABL kinase
inhibitor
is ponatinib. In certain preferred embodiments, the BCR-ABL kinase inhibitor
is a FLT3
inhibitor such as quizartinib (AC220), crenolanib (CP-868596), midostaurin
(PKC-
412), lestaurtinib (CEP-701), 4S0-203, TTT-3002, sorafenib (Bay-43-0006),
Ponatinib
(AP-24534), sunitinib (SU-11248), and/or tandutinib (MLN-0518), or (a)
pharmaceutically acceptable salt(s), solvate(s), and/or hydrate(s) thereof.
Preferably,
the FMS-like tyrosine kinase 3 (FLT3) inhibitor is quizartinib (AC220) or
pharmaceutically acceptable salt(s), solvate(s), and/or hydrate(s) thereof.
12
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
For instance, the topical formulation can have a mucosal surface residence
half-life on
esophageal tissue of at least 30 minutes, more preferably at least 60, 120,
180, 240
or even 300 minutes.
For instance, the topical formulation can produce at least a minimally
effective
concentration (MEC) of the ESO Regenerative Agent in the esophageal tissue to
which it is applied to which it is applied for at least 30 minutes, more
preferably at least
60, 120, 180, 240 or even 300 minutes.
For instance, the topical formulation can produce ESO Regenerative Agent
concentration in the esophageal tissue to which it is applied with T1/2 of at
least 2
hours, more preferably at least 4, 6, 8, 10 or even 12 hours.
In certain embodiments, the submucosal retentive formulation produces a
systemic
concentration of the ESO Regenerative Agent, such as ponatinib, which is less
than
1/3'd the maximum tolerated does (MTD) for that agent, and even more
preferably less
than 1/5th, 1/10th , 1/20th, 1/501h or even 1/100th the maximum tolerated does
(MTD) for
that agent.
In certain embodiments, the topical formulation is a viscous bioadhesive
liquid to coat
the esophagus.
In certain embodiments, the topical formulation comprises anti-PESC eluting
multiparticulates, microparticles, nanoparticles or microdiscs
In certain embodiments, the topical formulation further comprises an
anticoagulant.
In certain embodiments, the topical formulation further comprises comprises
one or
more antitussives, antihistamines, antipyretics, analgesics, anti-infective
agents
and/or chemotherapeutic agents
In further embodiments, there is provided bioadhesive nanoparticle having a
polymeric
surface with an adhesive force equivalent to an adhesive force of between 10
N/m2
and 100,000 N/m2 measured on human mucosal surfaces, which nanoparticle
further
includes at least one ESO Regenerative Agent, the ESO Regenerative Agent
dispersed therein or thereon, wherein the nanoparticle elutes the ESO
Regenerative
Agent into the mucous gel layer when adhered to mucosa! tissue. For example
the
formulation can include (i) a BCR-ABL kinase inhibitor, and (ii) one or more
pharmaceutically acceptable excipients, which formulation is injectable
submucosally
13
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
and forms a submucusal depot releasing an effective amount of the BCR-ABL
kinase
inhibitor to the surrounding tissue. In certain preferred embodiments, the BCR-
ABL
kinase inhibitor is ponatinib. In certain preferred embodiments, the BCR-ABL
kinase
inhibitor is a FLT3 inhibitor such as quizartinib (AC220), crenolanib (CP-
868596),
midostaurin (PKC-412), lestaurtinib (CEP-701), 48C-203, TTT-3002, sorafenib
(Bay-
43-0006), Ponatinib (AP-24534), sunitinib (SU-11248), and/or tandutinib (MLN-
0518),
or (a) pharmaceutically acceptable salt(s), solvate(s), and/or hydrate(s)
thereof.
Preferably, the FMS-like tyrosine kinase 3 (FLT3) inhibitor is quizartinib
(AC220) or
pharmaceutically acceptable salt(s), solvate(s), and/or hydrate(s) thereof.
In certain embodiments, the submucosal retentive formulation produces a
systemic
concentration of the ESO Regenerative Agent, such as ponatinib, which is less
than
1/3td the maximum tolerated does (MTD) for that agent, and even more
preferably less
than 1/51h, 1/101h , 1/20th, 1/50th or even 1/100th the maximum tolerated does
(MTD) for
that agent.
In certain embodiments, the bioadhesive nanoparticle further comprises an
anticoagulant.
In certain embodiments, the bioadhesive nanoparticle further comprises one or
more
antitussives, antihistamines, antipyretics, analgesics, anti-infective agents
and/or
chemotherapeutic agents
In further embodiments, there is provided a drug eluting device, which device
comprises drug release means including an anti-PESC agent, which device when
deployed in a patient positions the drug release means proximal to target
epithelial
tissue and releases the agent in an amount sufficient to achieve a
therapeutically
effective exposure of the target epithelial tissue to the agent.
In certain embodiments, the target epithelial tissue is esophageal tissue.
In certain embodiments, the target epithelial tissue is an epithelial-derived
tumor, such
as an ovarian tumor, a lung tumour, a gastric tumor or an esophageal tumor, or
a
metastatic site thereof
For instance, the drug eluting device can produce at least a minimally
effective
concentration (MEG) of the anti-PESC agent in the target epithelial tissue to
which it
14
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
is applied to which it is applied for at least 30 minutes, more preferably at
least 60,
120, 180, 240 or even 300 minutes.
For instance, the drug eluting device can produce anti-PESC agent
concentration in
the esophageal tissue to which it is applied with T1/2 of at least 2 hours,
more
preferably at least 4, 6, 8, 10 or even 12 hours.
In certain embodiments, the drug eluting device produces a systemic
concentration of
the anti-PESC agent which is less than 1/3rd the maximum tolerated does (MTD)
for
that agent, and even more preferably less than 1/5th, 1/10th , 1/20th, 1/50th
or even
1/1001h the maximum tolerated does (MTD) for that agent.
In certain embodiments, the drug eluting device is for treating one or more of
esophagitis, Barrett's esophagus, esophageal dysplasia or esophageal cancer,
which
device comprises drug release means including an Anti-BESC Agent that
selectively
kills or inhibits the proliferation or differentiation of Barrett's Esophagus
stem cells
(BESC) relative to normal esophageal stem cells, which device when deployed in
a
patient positions the drug release means proximal to the luminal surface of
the
esophagus and releases the agent in an amount sufficient to achieve a
therapeutically
effective exposure of the luminal surface to the agent.
Exemplary drug eluting devices include biodegradable stents, self-expandable
stents,
such as a self-expandable metallic stent (SEMS) or self-expandable plastic
stent
(SEPS), chips and wafers for submucusal implantation, and the like.
In other embodiments, the drug eluting device is a device for extraluminal
placement,
such as a microneedle cuff.
In certain embodiments, the anti-PESC agent is co-administered with an
analgesic,
and an anti-infective or both. These may be administered as separate
formulation, or
optionally, may be the anti-PESC agent is co-formulated with the analgesic or
the anti-
infective or both.
In certain embodiments, the anti-PESC agent is formulated as a liquid for oral
delivery
to the esophagus.
In certain embodiments, the anti-PESC agent is formulated as a single oral
dose.
In certain embodiments, the anti-PESC agent is delivered by a drug eluting
device that
is a drug eluting stent.
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In certain embodiments, the anti-PESC agent is delivered by a drug eluting
device that
is a balloon catheter having a surface coating including the agent.
In certain embodiments, the anti-PESC agent is cell permeable, such as
characterized
by a permeability coefficient of 10-8 or greater, more preferably 10-8 or
greater or 10-7
or greater.
In certain embodiments, the anti-PESC agent is a histone demethylase
inhibitor. In
certain preferred embodiments, the anti-PESC agent is a JmjC inhibitor. For
instance,
it can be a JmjC inhibitor that binds to and inhibits a catalytic JmjC domain.
In other
instances, it can be a JmjC inhibitor that is a plant homodomain (PHD)
inhibitor or a
protein-protein interaction inhibitor.
In certain embodiments, the anti-PESC agent is a pan-JmjC demethylase
inhibitor. An
exemplary JmjC inhibitor is JIB04. Other exemplary JmjC demethylase
inhibitors,
including pan-Jmje demethylase inhibitors, are described herein.
In certain embodiments, the anti-PESC agent is a receptor tyrosine kinase
inhibitor.
In certain preferred embodiments, the receptor tyrosine kinase inhibitor is an
EGFR
inhibitor, a HER2 inhibitor or a dual EGFR/HER2 inhibitor.
In certain embodiments, the anti-PESC agent is is a proteasome inhibitor.
In certain embodiments, the anti-PESC agent is a STAT inhibitor, and
preferably can
be a STAT3 inhibitor.
In certain embodiments, the anti-PESC agent is is a FLT3 inhibitor.
In certain embodiments, the anti-PESC agent is is a GSK3 inhibitor.
In certain embodiments, the anti-P ESC agent is is a HSP90 inhibitor, a HSP70
inhibitor
or a dual HSP90/HSP70 inhibitor.
In certain embodiments, the anti-PESC agent is a selected from the group
consisting
of
16
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
0
0 0
>rit'00-
oF10., B.,OH
0
----
....N I H
r)
H N
,LtNs..
Delanzomib (CEP-18770);
F121.1
Adefovir Dipivoxyl;
ri
01
i=i% 411 \ o
H
NH
,....., -,..N ... N a
\ /
_.-e
CI
Nnoi
JIB04;
AZD1080.
('1\1"-
0 0 0
CI
g
0 . N)
e,
4-- N 0- so
---"----AN . Srsi: AN 1
/
H H
S
VVZ8040;
tattic;
....,0
210.4.0
OH
HO--',.. .õ..
0
- CI
-;
= = =
H
Nanchangnnycin; and
In certain embodiments of the methods, preparations and devices of the present
disclosure the anti-PESC agent is administered with a second drug agent that
selectively promotes proliferation or other regenerative and wound healing
activities
of normal epithelial stem cells (an "ESO Regenerative agent") with an EC50 at
least 5
times more potent than for PESCs, more preferably with an ECso 10 times, 50
times,
17
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
100 times or even 1000 times more potent for normal epithelial stem cells
relative to
for PESCs.
In certain embodiments of the methods, preparations and devices of the present
disclosure the anti-PESC agent is administered with a second drug agent that
selectively promotes proliferation or other regenerative and wound healing
activities
of normal esophageal stem cells (an "esophageal ESO Regenerative agent") with
an
EC50 at least 5 times more potent than for BESCs, more preferably with an EC50
10
times, 50 times, 100 times or even 1000 times more potent for normal
esophageal
stem cells relative to for BESCs.
In certain embodiments of the methods, preparations and devices of the present
disclosure the anti-PESC agent is administered with an ESO Regenerative agent
that
selectively promotes proliferation of normal epithelial stem cells with an
ECso of 10-8
M or less, more preferably 10-7 M or less, 10-8 M or less or 100 M or less.
In certain embodiments of the methods, preparations and devices of the present
disclosure the anti-PESC agent is administered with an esophageal ESO
Regenerative agent that selectively promotes proliferation of normal
esophageal stem
cells with an EC50 of 10-8 M or less, more preferably 10-7 M or less, 10-8 M
or less or
10-9 M or less.
In certain embodiments, the combined administration of the anti-PESC agent and
the
ESO Regenerative agent has a therapeutic index (1-1) for treating esophagitis,
Barrett's
Esophagus, esophageal dysplasia and/or esophageal cancer of at least 2, and
more
preferably has a therapeutic index of at least 5, 10. 20. 50. 100, 250. 500 or
1000 for
treating esophagitis, Barrett's Esophagus, esophageal dysplasia and/or
esophageal
cancer.
In certain embodiments, the combined administration of the anti-PESC agent and
the
ESO Regenerative agent has a therapeutic index (TI) for treating ovarian,
fallopian
and or cervical metaplasia or dysplasia of at least 2, and more preferably has
a
therapeutic index of at least 5, 10, 20, 50, 100, 250, 500 or 1000.
In certain embodiments, the combined administration of the anti-PESC agent and
the
ESO Regenerative agent has a therapeutic index (TI) for treating ovarian
cancer (such
as taxol and/or cisplatin resistant ovarian cancer) of at least 2, and more
preferably
has a therapeutic index of at least 5, 10, 20, 50, 100, 250, 500 or 1000.
18
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In certain embodiments, the combined administration of the anti-PESC agent and
the
ESO Regenerative agent has a therapeutic index (TI) for treating lung cancer
(such
NSCLC or SCLC) of at least 2, and more preferably has a therapeutic index of
at least
5, 10, 20, 50, 1001 250, 500 or 1000.
In certain embodiments, the combined administration of the anti-PESC agent and
the
ESO Regenerative agent has a therapeutic index (TI) for treating lung
metaplasia or
dysplasia of at least 2, and more preferably has a therapeutic index of at
least 5, 10,
20, 50, 100, 250, 500 or 1000.
In certain embodiments, the ESO Regenerative agent is pan-inhibitor of ABL
kinase
inhibitor, preferably a BCR-ABL kinase inhibitor Exemplary pan-inhibitor
include
imatinib, nilotinib, dasatinib, bosutinib and ponatinib, and is preferably
ponatinib.
In certain embodiments, the ESO Regenerative agent is a tyrosine kinase
inhibitor,
which at its Cmax concentration following its route of administration,
inhibits one or
more of FLT3, Bcr-Abl, c-KIT, PDGFR, VEGFR and FGFR with a Ki for inhibition
that
is less than 1/2 the EC50 for slowing or reversing the progress of an
esophageal
metaplasia, dysplasia, cancer or a combination thereof, and even more
preferably
115th, 1/10th. 1/20th , 1/50th or even 1/100th .
In certain embodiments, the anti-PESC agent and the ESO Regenerative agent are
administered to the patient as separate formulations.
In certain embodiments, the anti-PESC agent and the ESO Regenerative agent are
co-formulated together
One aspect of the disclosure provides a single oral dosage formulation
comprising (i)
an anti-PESC agent, (ii) an ESO Regenerative Agent, and (iii) and a
pharmaceutically
acceptable excipient, which single oral dosage formulation taken by an adult
human
patient produces a concentration of anti_PESC agent and ESO Regenerative Agent
in esophageal tissue effective to slow or reverse the progress of an
esophageal
metaplasia, dysplasia, cancer or a combination thereof In certain preferred
embodiments, the BCR-ABL kinase inhibitor is ponatinib. In certain preferred
embodiments, the BCR-ABL kinase inhibitor is a FLT3 inhibitor such as
quizartinib
(AC220), crenolanib (CP-868596), midostaurin (PKC-412), lestaurtinib (CEP-
701),
4SC-203, TTT-3002, sorafenib (Bay-43-0006), Ponatinib (AP-24534), sunitinib
(SU-
11248), and/or tandutinib (MLN-0518), or (a) pharmaceutically acceptable
salt(s),
19
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
solvate(s), and/or hydrate(s) thereof. Preferably, the FMS-like tyrosine
kinase 3 (FLT3)
inhibitor is quizartinib (AC220) or pharmaceutically acceptable salt(s),
solvate(s),
and/or hydrate(s) thereof.
In certain embodiments, the methods, preparations and devices of the present
disclosure are intended (and appropriate) for use in human patients.
Still another aspect of the present disclosure provides a method for treating
a patient
suffering from one or more of esophagitis, Barrett's Esophagus, esophageal
dysplasia
or esophageal cancer, which method comprises administering to the esophagus of
a
patient a nucleic acid construct that reduces the level of expression of one
or more of
JrnjC, EGFR, HER2, proteasome subunits, STAT3, FLT3, GSK3, HSP9Oand/or
HSP70 to selectively kill or inhibit the proliferation or differentiation of
Barrett's
Esophagus stem cells (BESC) relative to normal esophageal stem cells.
Exemplary
nucleic acid constructs include RNAi constructs (nucleic acids which reduce
expression through an RNA interference mechanism) such as siRNA, shRNA or
miRNA, as well as antisense nucleic acids. In certain embodiments, the patient
presents with esophagitis. In certain embodiments, the patient presents with
Barrett's
Esophagus. In certain embodiments, the patient presents with esophageal
dysplasia.
In certain embodiments, the patient presents with esophageal cancer. In
certain
embodiments, the patient presents with esophageal carcinoma, such as
esophageal
adenocarcinoma or esophageal squamous cell carcinoma.
Similarly, another aspect of the present disclosure provides a method for
treating a
patient suffering from one or more of esophagitis, Barrett's Esophagus,
esophageal
dysplasia or esophageal cancer, which method comprises administering to the
esophagus of a patient a CRISPR construct that reduces the level of expression
of
one or more of JnnjC, EGFR, HER2, proteasonne subunits, STAT3, FLT3, GSK3,
HSP9Oand/or HSP70 to selectively kill or inhibit the proliferation or
differentiation of
BESCs relative to normal esophageal stem cells. In certain embodiments, the
patient
presents with Barrett's Esophagus. In certain embodiments, the patient
presents with
esophageal dysplasia. In certain embodiments, the patient presents with
esophageal
cancer. In certain embodiments, the patient presents with esophageal
carcinoma,
such as esophageal adenocarcinoma or esophageal squamous cell carcinoma.
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Description of the Fit:lures
Figures 1A-G. Establishment of HTS screening on BE stem cells.
Figure -1A. Schematic of chemical screening pipeline. 1mm biopsy of BE lesion
was processed and mixed BE stem cells (Krt7+) and ESO stem cell (Krt5+)
were cloned and culture in StemECHO cell culture system. Single cell derived
pedigrees of Krt5+ ESO stem cells and Krt7+ BE stem cells were established,
labeled with GFP and subject to chemical screening. Drugs that eradicate the
BE stem cells were identified as hits.
Figure 113. Summary of the chemical libraries being used for the screenings.
Figure 1C. Sensitivity clustering of cell lines used in the screening. BE stem
cells and ESO stem cells showed overall distinct sensitivity to chemicals
included in the screenings.
Figure 1D. Left, Scatter plot comparing median survival rate of BE stem cells
and ESO stem cells. Right, Ponatinib promoted the proliferation of ESO stem
cells.
Figure 1E. Left, Representative images of ESO and BE stem cells in the
absence or presence of Ponatinib. Right, Dosage response of BE and ESO
stem cells towards Ponatinib.
Figure IF. Representative image of a scanned 384 plate showing the effects
of the chemicals on GFP labeled stem cells.
Figure 1G. Example of converting green binary overlay to the heatmap
representing the total area of colonies in each individual well.
Figures 2A-F. Synergistic HTS screening to identify dual-function drug
combinations.
Figure 2A. Scatter plot display of the HTS result of synergistic HTS. Selected
eight top hits were highlighted in red.
Figure 2B. Summary of the information of selected top hits.
21
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Figure 2C. Dosage response curves of the top eights hits validated their
differential effects to BE and ESO stem cells.
Figure 20. Left, Scanned images of BE (Krt7+, Green)/ESO (Krt5+, Red) co-
culture system for the validation of seven selected drugs on eight patients.
Right, Quantitative analysis of the scanned images.
Figure 2E. Scanned image of GFP labeled BE stem cells treated with a
gradient of selected hits from HTS.
Figure 2F. Dosage curves of the hits showed that all selected ones were
validated to eradicate BE stem cells. Numeric values obtained from GFP signal
threshold image analysis were used to generate dose response curves.
Figures 3A-E. Validation of selected hits in 3D and mouse models.
Figure 3A. Upper, Schematic of the 3D culture validation. Lower,
Representative images of top view and histological sections of untreated and
treated 3D co-culture structures. BE stem cells (Krt7+, Green) and ESO stem
cells (Krt5+, Red) co-existed in the absence of selected drug combination.
Following the treatment, BE stem cells (Green) were eradicated while ESO
stem cells (Red) compensated the blanked area. Figure 3B. Quantitative
analysis of 3D co-culture treatment images of top view from eight patients_
Figure 3C. Schematic of drug combination validation in a mouse model. Mice
were co-injected subcutaneously with a mixture of BE (GFP labeled) and ESO
stem cells. Following the serial treatments, the GFP+ BE stem cells became
invisible.
Figure 3D. Histological analysis of BE stem cells (Krt7+, Green)/ESO stem
cells (Krt5+, Red) co-transplanted structures from treated and non-treated
mice.
Figure 3E. Quantitative analysis of clonogenic assay performed by processing
the untreated and treated transplants and culturing in StemECHO culture
system.
Figure 3F. Scanned images of BE stem cells and ESO stem cells treated with
a range of dosages of eight selected hits together with 1 pM Ponatinib. Their
22
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
differential effects on BE and ESO stem cells were validated except AZD1080
that appeared to be the false positive hit in the screening.
Figures 4A-E. Drug combinations eradicated patient-matched BE, dysplasia and
cancer.
Figure 4A. PCA map of stem cell gene expression from Barrett's, dysplastic,
tumor and normal esophagus.
Figure 4B. Venn diagrams of overlapped genes among BE, dysplasia and
cancer stem cells versus ESO stem cells.
Figure 4C. Common pathways enriched in BE, dysplasia and cancer.
Figure 4D. Pathways targeted by the hits identified in HTS.
Figure 4E. Upper, Scanning images of co-cultured BE stem cells (Krt7+,
Green) and ESO stem cells (Krt5+, Red) treated with seven different drug
combinations. Lower, Quantitative analysis of the scanned images showed that
patient-matched BE, dysplasia and cancer stem cells could be eradicated with
the same drug combinations.
Figures 5A-G. CEP-18770 and JIB04 together with Ponatinib eradicated BE,
dysplasia and cancer stem cells in vitro and in viva
Figure 5A. Schematic of 3D culture validation on patient-matched BE,
dysplasia and cancer stem cells.
Figure 5B. Left, Scanned images of top view of BE, dysplasia and cancer stem
cells (green) generated ALI structures in the presence of absence of drug
combinations. Right, Quantitative analysis of the images of ALI structures
showed that CEP-18770 and JIB04 combined with Ponatinib eradicated BE,
dysplasia and cancer stem cells.
Figure 5C. Schematic of in vivo validation of drug combinations in mice
xenografted with dysplasia and cancer stem cells. Following the treatment, the
xenografts were processed for histological analysis or clonogenic assays to
detect the existence of stem cells.
23
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Figure 50. Dysplastic structures (Krt7+, Green/Ki67+, Red) were diminished
following CEP-19770/Ponatinib or JIB04/Ponatinib treatment. Consistently,
stem cells were not cloned from treated structures as shown in scanned
rhodamine staining.
Figure 5E. Cancer structures (Krt7+, Ki67+) were diminished following the drug
treatment. Rhodamine staining showed the cancer stem cells were not cloned
from the treated structures.
Figures 5F and 5G. BE stem cells (Krt7+, Green) and ESO stem cells (Krt5+,
Red) were co-injected into the NSG mice and treated with JIB04 or CEP-18770
with Ponatinib. The tissues were collected and fixed for histological
analysis.
BE stem cells were not detected in the treated structures while the Krt5+ ESO
structures were visibly more robust in the treated samples.
Figure 6. Patient-matched BE, dysplasia and cancer stem cells (Krt7+, Green)
were
mixed with Krt5+ (red) ESO cells and co-cultured for five days and then
treated with
seven selected drugs together with 1 pM Ponatinib. Scanning image and
quantitative
analysis showed that all seven drug combinations reduced the growth of BE,
dysplasia
and cancer stem cell growth, while JIB04 and CEP-18770 remained the most
effective
drugs.
Figures 7A-7D. Exemplary classes and illustrative structures of histone
deacetylase
inhibitors useful in the present disclosure.
Figures 8. Is a diagram representing the continuum in certain epithelial
tissues of
metaplasia to dysplasia to cancer.
Figures 9. Is a diagram showing the statistically increasing risk of a patient
developing
esophageal adenocarcinoma as disease progresses from Barrett's esophagus to
high
grade dysplasia.
Figures 10A. Demonstrates the synergy in the combination of JIB04 with
ponatinib
in killing pathogenic BE stem cells relative to normal esophageal stem cells.
Projected
therapeutic index is greater than 200 fold.
Figures 106. Demonstrates the synergy in the combination of JIB04 with
ponatinib
in killing both BE stem cells, as well as stem cells from biopsies graded as
dysplasia
24
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
and Esophageal Adenocarcinoma (EAC). Projected therapeutic index is greater
than
100 fold.
Figures 11A. Pathogenic stem cells isolated from high grade ovarian tumor
biopsies
obtained from drug naïve patients already include cisplatin-resistant stem
cell
populations. PCA analysis of cisplatin-resistant stem cell populations from
drug naive
patients indicates that these cells cluster with pathogenic stem cells from
stem cells
isolated from cisplatin-resistant tumors from patients having relapsed or
failed cisplatin
therapy.
Figures 11B. Demonstrates the synergy in the combination of taxol with
ponatinib in
killing taxol-resistant ovarian stem cells in vitro.
Figures 11C. Demonstrates the synergy in the combination of taxol with
ponatinib in
reducing tumor volume taxol-resistant ovarian stem cells in xenograft animal
models.
Figures 12. Pathogenic stem cells can be cloned and cultured from lung cancers
(both small cell and non-small cell lung cancer). These pathogenic stem cells,
along
with normal regenerative lung stem cells, can be adapted to high throughput
screening
plates.
Figures 13. Demonstrates the synergy in the combination of JIB04 with
ponatinib in
killing both NSCLC and SCLC stem cells relative to normal lung epithelial stem
cells.
Description of Certain Embodiments
I. Overview
Barrett's Esophagus holds a pivotal position at the interface of cancer
biology and
patient care. Barrett's was first discovered in 1950's and associated with
risk for
adenocarcinoma in the 1970's. Barrett's has become a paradigm for precancerous
lesions giving rise to progressively more advanced lesions in a process
requiring many
years supporting an overall escalation model whereby non-cancerous lesions
undergo
long-term processes of stochastic changes some of which yield more sinister
and
determinant transitions to low- and high-grade dysplasia which then rapidly
and almost
inexorably evolve to malignant disease. The recognition of the importance of
preemptive therapies that target these prennalignant lesions is the foundation
of cancer
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
prevention. If true, the clinical solution to preventing the onset of
esophageal
adenocarcinoma would be simple and direct: ablate Barrett's before it can
evolve to
more aggressive lesions.
The advance of the development of targeted therapies for Barrett's requires
conceptual advance of the origin of Barrett's and the recognition of the
existence of
Barrett's stem cells. If the premalignant stages of EAC represent the only
tractable
solution to this disease, it is essential to solve the mystery of the origin
of BE and
develop new therapeutic strategies specifically targeting its stem cells.
However, the
ontogeny of BE has been an intriguing puzzle with various hypotheses involving
transconnnnitnnent of esophageal squannous stem cells, migration from lower
gastrointestinal sites, the reparative emergence of subnnucosal glands,
dissemination
from bone marrow. We recently showed that BE originated from the opportunistic
growth of residual embryonic cells pre-existing at gastroesophageal junction
(Wang et
al, Cell. 2011 Jun 24;145(7):1023-1035). In addition, using the ground state
stem cell
technology that enabled us to clone stem cells of the normal human
gastrointestinal
tract, we demonstrated the existence of the stem cells in BE (Yamamoto at at,
Nat
Commun. 2016 Jan 19;7:10380) and suggested they are the key elements to target
in
a therapeutic program designed to prevent the development and progression of
this
irreversible and dangerous metaplasia.
In order to uncover drugs specifically targeting BE stem cells that might
synergize with
physical ablation protocols to further reduce recurrent disease, provided
herein is a
multiplexed screening of established and experimental drugs or combinations
thereof
to identify compounds and combinations of compounds that selectively target
the
particular pathways that dominate the survival of these BE lesions. These BE
stem
cells were used in hybrid models with normal epithelial squamous stem cells to
model
the potential ability of such drug combinations to alter the competitive
status of such
lesions in the distal esophagus.
Also provided herein are screening methods that show the similar selective
vulnerabilities of the stem cells of patient-matched BE, dysplasia and EAC,
which
suggest the broad usage of the pharmacological compositions that would augment
physical ablation or mucosal dissection therapies. Indeed, as demonstrated by
the
data presented herein, the differential sensitivity of the pathogenic stem
cells to single
26
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
agents or combination therapies is carried across multiple tissues and across
metaplasia, dysplasia or tumor samples from those tissues.
II. Definitions
Unless otherwise stated, the following terms used in the specification and
claims are
defined for the purposes of this Application and have the following meaning:
"Alkyl" means a linear saturated monovalent hydrocarbon radical of one to six
carbon
atoms or a branched saturated monovalent hydrocarbon radical of three to six
carbon
atoms, e.g., methyl, ethyl, propyl, 2-propyl, butyl (including all isomeric
forms), pentyl
(including all isomeric forms), and the like. The term "alkyl," as used
herein, in
synonymous with the term "aliphatic."
The term "alkenyl" as used herein describes groups which are preferably
lower alkenyl containing from two to eight carbon atoms in the principal chain
and up
to 20 carbon atoms. They may be straight or branched chain or cyclic and
include
ethenyl, propenyl, isopropenyl, butenyl, isobutenyl, hexenyl, and the like.
The term "alkynyl" as used herein describes groups which are preferably lower
alkynyl
containing from two to eight carbon atoms in the principal chain and up to 20
carbon
atoms. They may be straight or branched chain and include ethynyl, propynyl,
butynyl,
isobutynyl, hexynyl, and the like.
"Alkylsulfonyl" means a -SO2R radical where R is alkyl as defined above, e.g.,
methylsulfonyl, ethylsulfonyl, and the like.
"Alkoxy" means an -OR radical where R is alkyl as defined above, e.g.,
methoxy,
ethoxy, propoxy, or 2-propoxy, n-, iso-, or tert-butoxy, and the like.
"Aminoalkyl" means a linear monovalent hydrocarbon radical of one to six
carbon
atoms or a branched monovalent hydrocarbon radical of three to six carbons
substituted with at least one, preferably one or two, -NRR where R is
hydrogen, alkyl,
or -CORa where Ra is alkyl, each as defined above, and R is selected from
hydrogen, alkyl, hydroxyalkyl, alkoxyalkyl, aryl, aralkyl, heteroaryl,
heteroaralkyl, or
haloalkyl, each as defined herein, e.g., aminomethyl, methylaminoethyl, 2-
ethylamino-
2-methylethyl, 1, 3-diaminopropyl, dim ethylam inomethyl,
diethylam inoethyl,
acetylaminopropyl, and the like.
27
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
"Aminosulfonyl" means a -SO2NRIR' radical where R is independently hydrogen,
alkyl,
hydroxyalkyl, alkoxyalkyl, or aminoalkyl, each as defined herein and R' is
hydrogen, alkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heteroaryl,
heteroaralkyl,
heterocyclyl, heterocyclylalkyl, hydroxyalkyl, alkoxyalkyl, or aminoalkyl,
each as
defined herein, e.g., - 802NH2, methylaminosulfonyl, 2-dimethylaminosulfonyl,
and the
like.
"Acyl" means a -COR radical where R is alkyl, haloalkyl, cycloalkyl,
cycloalkylalkyl, aryl, aralkyl, heteroaryl,
heteroaralkyl, heterocyclyl, or
heterocyclylalkyl, each as defined herein, e.g., acetyl, propionyl, benzoyl,
pyridinylcarbonyl, and the like. When R is alkyl, the radical is also referred
to herein
as alkylcarbonyl.
"Acylamino" means an -NHCOR radical where R is alkyl, haloalkyl, cycloalkyl,
cycloalkylalkyl, aryl, aralkyl, heteroaryl,
heteroaralkyl, heterocyclyl, or
heterocyclylalkyl, each as defined herein, e.g., acetylamino, propionylamino,
and the
like.
"Aryl" means a monovalent monocyclic or bicyclic aromatic hydrocarbon radical
of 6
to 10 ring atoms e.g., phenyl or naphthyl. As used herein, "aryl" and
"aromatic" may
be used interchangeably.
"Bridged heterocyclyl" means a saturated or unsaturated monovalent bicyclic
group of
5 to 10 ring atoms in which one or two ring atoms are heteroatom selected from
N, 0,
or S(0)n where n is an integer from 0 to 2, the remaining ring atoms being C,
where
some of the rings are created by one or more bridges.
"Cycloalkyl" means a cyclic, saturated, monovalent hydrocarbon radical of
three to ten
carbon atoms, e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl, and
the like.
The cycloalkyl ring can optionally be fused to phenyl or monocyclic heteroaryl
ring as
defined herein. When the cycloalkyl ring is referred to herein as 'fused
cycloalkyl", it
means that the cycloalkyl ring is fused to phenyl or monocyclic heteroaryl
ring. When
the cycloalkyl ring is referred to herein as "monocyclic cycloalkyl", it means
that the
cycloalkyl ring is not fused to phenyl or monocyclic heteroaryl ring. As used
herein,
"cycloalkyl," "carbocycle," and "carbocyly1" may be used interchangeably.
"Carboxy" means -COON.
28
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
"Disubstituted amino" means an -NRR' radical where R and R' are independently
alkyl,
cycloalkyl, cycloalkylalkyl, acyl, sulfonyl, aryl, aralkyl, heteroaryl,
heteroaralkyl,
heterocyclyl, heterocyclylalkyl, hydroxyalkyl, alkoxyalkyl, or aminoalkyl,
each as
defined herein, e.g., dimethylamino, phenylmethylamino, and the like. When R
and R'
are alkyl, the group is referred to herein as dialkylamino
"Halo" means fluoro, chloro, bromo, or iodo, preferably fluoro or chloro.
"Haloalkyl"
means alkyl radical as defined above, which is substituted with one or more
halogen
atoms, preferably one to five halogen atoms, preferably fluorine or chlorine,
including
those substituted with different halogens, e.g., -CH2C1, -CF3, -CHF2, -CH2CF3,
-
CF2CF3, -CF(CH3)2, and the like. When the alkyl is substituted with only
fluoro, it is
referred to in this Application as fluoroalkyl.
"Hydroxyalkyl" means a linear monovalent hydrocarbon radical of one to six
carbon
atoms or a branched monovalent hydrocarbon radical of three to six carbons
substituted with one or two hydroxy groups, provided that if two hydroxy
groups are
present they are not both on the same carbon atom. Representative examples
include,
but are not limited to, hydroxym ethyl, 2-hydroxy ethyl, 2-
hydroxypropyl, 3-
hydroxypropyl, 1-(hydroxymethyl)-2- methylpropyl, 2-hydroxybutyl, 3-
hydroxybutyl, 4-
hydroxybutyl, 2,3-dihydroxypropyl, 1- (hydroxymethyl)-2-hydroxyethyl, 2,3-
dihydroxybutyl, 3,4-dihydroxybutyl and 2- (hydroxymethyl)-3-hydroxypropyl,
preferably 2-hydroxyethyl, 2,3-dihydroxypropyl, or 1- (hydroxymethyl)-2-
hydroxyethyl.
"Heterocycly1" means a saturated or unsaturated monovalent monocyclic group of
4
to 8 ring atoms in which one or two ring atoms are heteroatom selected from N,
0, or
S(0)n, where n is an integer from 0 to 2, the remaining ring atoms being C.
The
heterocyclyl ring is optionally fused to a (one) aryl or heteroaryl ring as
defined herein
provided the aryl and heteroaryl rings are monocyclic. The heterocyclyl ring
fused to
monocyclic aryl or heteroaryl ring is also referred to in this Application as
"bicyclic
heterocyclyl" ring. Additionally, one or two ring carbon atoms in the
heterocyclyl ring
can optionally be replaced by a -CO- group. More specifically the term
heterocyclyl
includes, but is not limited to, pyrrolidino, piperidino, homopiperidino, 2-
oxopyrrolidinyl,
2-oxopiperidinyl, morpholino, piperazino, tetrahydropyranyl, thiomorphol ino,
tetrahydroisoquinolinyl, and the like. When the heterocyclyl ring is
unsaturated it can
contain one or two ring double bonds provided that the ring is not aromatic.
When the
heterocyclyl group contains at least one nitrogen atom, it is also referred to
herein as
29
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
heterocycloamino and is a subset of the heterocyclyl group. When the
heterocyclyl
group is a saturated ring and is not fused to aryl or heteroaryl ring as
stated above, it
is also referred to herein as saturated monocyclic heterocyclyl.
"Heteroaryl" means a monovalent monocyclic or bicyclic aromatic radical of 5
to 10
ring atoms where one or more, preferably one, two, or three, ring atoms are
heteroatom selected from N, 0, or S, the remaining ring atoms being carbon.
Representative examples include, but are not limited to, pyrrolyl, thienyl,
thiazolyl,
imidazolyl, furanyl, indolyl, isoindolyl, oxazolyl, isoxazolyl,
benzothiazolyl,
benzoxazolyl, quinolinyl, isoquinolinyl, pyridinyl, pyrimidinyl, pyrazinyl,
pyridazinyl,
triazolyl, tetrazolyl, and the like.
"Monosubstituted amino" means an -NHR radical where R is alkyl, cycloalkyl,
cycloalkylalkyl, acyl, sulfonyl, aryl, aralkyl, heteroaryl, heteroaralkyl,
heterocyclyl,
heterocyclylalkyl, hydroxyalkyl, alkoxyalkyl, or aminoalkyl, each as defined
herein,
e.g., methylamino, 2-phenylamino, hydroxyethylamino, and the like. When R is
alkyl,
the group is referred to herein as monoalkylamino.
A "pharmaceutically acceptable salt" of a compound means a salt that is
pharmaceutically acceptable and that possesses the desired pharmacological
activity
of the parent compound. Such salts include: acid addition salts, formed with
inorganic
acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid,
phosphoric
acid, and the like; or formed with organic acids such as formic acid, acetic
acid,
propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid,
pyruvic acid,
lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric
acid, tartaric
acid, citric acid, benzoic acid, 3-(4- hydroxybenzoyl)benzoic acid, cinnamic
acid,
mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1 ,2-
ethanedisulfonic acid,
2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic
acid, 2-
naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid,
glucoheptonic
acid, 4,4'-nnethylenebis-(3-hydroxy-2-ene-l-carboxylic acid), 3-
phenylpropionic acid,
trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid,
gluconic acid,
glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic
acid, and
the like; or salts formed when an acidic proton present in the parent compound
either
is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion,
or an
aluminum ion; or coordinates with an organic base such as ethanolamine,
diethanolamine, triethanolamine, tromethamine, N-methylglucamine, and the
like. It is
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
understood that the pharmaceutically acceptable salts are non-toxic.
Additional
information on suitable pharmaceutically acceptable salts can be found in
Remington's
Pharmaceutical Sciences, 17th ed., Mack Publishing Company, Easton, PA, 1985,
which is incorporated herein by reference. The compounds of the present
disclosure
can also exist as cocrystals.
The compounds of the present disclosure may have asymmetric centers. Compounds
of the present disclosure containing an asymmetrically substituted atom may be
isolated in optically active, racemic forms or other mixtures of isomers. It
is well known
in the art how to prepare optically active forms, such as by resolution of
materials. All
chiral, diastereonneric, racennic forms are within the scope of this
disclosure, unless
the specific stereochennistry or isomeric form is specifically indicated.
Certain compounds of can exist as tautomers and/or geometric isomers. All
possible
tautomers and cis and trans isomers, as individual forms and mixtures thereof
are
within the scope of this disclosure. Additionally, as used herein the term
alkyl includes
all the possible isomeric forms of said alkyl group albeit only a few examples
are set
forth.
A "pharmaceutically acceptable carrier or excipient" means a carrier or an
excipient
that is useful in preparing a pharmaceutical composition that is generally
safe, non-
toxic and neither biologically nor otherwise undesirable, and includes a
carrier or an
excipient that is acceptable for veterinary use as well as human
pharmaceutical use.
"A pharmaceutically acceptable carrier/excipient" as used in the specification
and
claims includes both one and more than one such excipient.
"Sulfonyl" means a -SO2R radical where R is alkyl, haloalkyl, aryl, aralkyl,
heteroaryl,
heteroaralkyl, heterocyclyl, heterocyclylalkyl, each as defined herein, e.g.,
methylsulfonyl, phenylsulfonyl, benzylsulfonyl, pyridinylsulfonyl, and the
like. When R
is alkyl, it is also referred to herein as alkylsulfonyl.
"Substituted alkyl" means a linear saturated monovalent hydrocarbon radical of
one to
six carbon atoms or a branched saturated monovalent hydrocarbon radical of
three to
six carbon atoms where one or two hydrogen atoms in the alkyl chain are
independently replaced by hydroxyl, halo, alkoxy, amino, monosubstituted
amino,
disubstituted amino, cyano, sulfonyl, aminocarbonyl, aminosulfonyl, -NHCONH2,
carboxy, acyl, acylamino, phenyl, or alkoxycarbonyl, each group as defined
herein.
31
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
"Substituted alkynyl" means a linear saturated monovalent hydrocarbon radical
of two
to six carbon atoms or a branched monovalent hydrocarbon radical of three to
six
carbon atoms containing a triple bond where one or two hydrogen atoms in the
alkynyl
chain are independently replaced by phenyl, hydroxyl, alkoxy, amino,
monosubstituted
amino, disubstituted amino, cyano, sulfonyl, aminocarbonyl, am inosulfonyl, -
NHCONH2, carboxy, acyl, acylamino, or alkoxycarbonyl, each group as defined
herein.
"Treating" or "treatment" of a disease includes: preventing the disease, i.e.
causing
the clinical symptoms of the disease not to develop in a mammal that may be
exposed
to or predisposed to the disease but does not yet experience or display
symptoms of
the disease; inhibiting the disease, La, arresting or reducing the development
of the
disease or its clinical symptoms; or relieving the disease, i.e., causing
regression of
the disease or its clinical symptoms.
III. Exemplary Embodiments
a. Histone Demethylase Inhibitors
The Jumonji family of histone demethylases has been the focus of much study
over
the last few years. Numerous reports have demonstrated the relevance of these
enzymes in a variety of physiological and pathological conditions beyond
cancer,
including early development, reproduction, metabolism, and cardiac
hypertrophy. The
structure of Jumonji catalytic domains shows they are drugable, able to
accommodate
small molecule disruptors, making them ideal molecular targets for
intervention.
Modulation of aberrant Jumonji demethylase activity in disease should lead to
the
normalization of transcriptional patterns, such as we see with JI1304 in
cancer cells.
JIB04's ability to block tumor growth and prolong cancer survival may involve
both
direct and indirect aggregate effects of Jumonji enzyme pan-inhibition in
cells and in
vivo, and it is possible that the drug accumulate in cancer cells over time
increasing
its effective concentration/apparent potency. Mechanistically, JIB04 appears
to
chelate iron in the catalytic site of Jumonji enzymes and to disrupt histone
substrate
binding, while not being a competitive inhibitor for a-ketoglutarate, a
mechanism not
yet described for Jumonji inhibitors. Future structural information will be
necessary to
establish the exact molecular interactions between Jumonji enzymes and their
32
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
cofactors/substrates that are disrupted by JIB04. One possibility is that the
inhibitor
may occupy the outer portion of the active site where the iron and the histone
substrate
bind or bind iron in the active site in a manner that alters subsequent
substrate binding.
Chelation in solution may also contribute to inhibition under conditions of
high free
iron.
In certain embodiments, the anti-PESC agent is JIB-004
r.:11
NH
-
ci
JIB04 (NSC693627, E-isomer) is a potent, selective and cell permeable Jumonji
histone demethylase inhibitor. Unlike the other known inhibitors, JIB04 is not
a
competitive inhibitor of a-ketoglutarate. It inhibits the demethylase activity
of Jumonji
enzymes in vitro, with ICso -230 nM for JARID1A (KDM5A), -440 nM for JMJD2A
(KDM4A) and JMJD2B (KDM4B), -340 nM for JMJD2E (KDM2E), and -1 pM for
JMJD3 (KDM6B) and JWIJD2C (KDM4C). JIB04 blocks Jumonji demethylase activity
in cells and consequently inhibits cell growth, without affecting other a-
ketoglutarate-
dependent hydroxylases or histone-modifying enzymes, especially HDACs. JIB04
alters transcriptional programs in cancer but not in normal cells, leading to
cancer-
specific cell death. Importantly, in vivo, JIB04 lowers histone demethylase
activity in
tumors, reduces tumor burden and prolongs survival of mice in an aggressive
breast
cancer model.
In some embodiments, the agent that inhibits a JmjC polypeptide is JIB04, SD-
70,
ML324, KDM5-C70, PBIT, KDOHP64a, KDOQZ5, 10X1, 10X2, KDOMA83,
KDMOBP69, N5C636819, pyrido[3,4-pyrimidin-4(3H)-one derivatives, 3- amino-4-
pyridine carboxylate derivatives or analogs thereof. See PCT W02017190009A1
ML-324
33
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
OH
ii
JmjC Histone Demethylase Inhibitor, n-Octy1-10X1
0 0
1;
k
r."
6H
KDM4A/KDM4B Inhibitor, NSC636819
oti,rst-õõ woe
'c---
i
CA '
JMJD2 Inhibitor, 5-carboxy-8HQ - CAS 5852-78-8
0 OH
µN-?
OH
JMJD Histone Demethylase Inhibitor III ¨ Calbiochem
34
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
4444
i ?*1
JMJD Histone Demethylase Inhibitor IV, Methylstat - CAS 1310877-95-2 ¨
Calbiochem
753
JIB04 as well as other suitable histone demethylase inhibitors are also
described in
Thinnes at at Biochimica et Biophysica Acta - Gene Regulatory Mechanisms,
Volume
1839, Issue 12, December 2014, Pages 1416-1432 and Epigenetic Drug Discovery
ed. Wolfgang Sippl, Manfred Jung, Raimund Mannhold, Helmut Buschmann, JOrg
Holenz, John Wiley & Sons, Feb 11, 2019 ISBN: 978-3-527-34314-0 - (both of
which
are incorporated by reference herein).
In one embodiment, the JMJD3 demethylase inhibitor comprises ethyl 3-((6-(4,5-
dihydro-1H-benzo[d]azepin-3(2H)-y1)-2-(pyridin-2-yl)pyrim
ino)propanoate
(GSK-J4), and active derivatives thereof as disclosed in Kruidenier et at, "A
Selective
Jumonji H3K27 Demethylase Inhibitor Modulates Proinflammatory Macrophage
Response," Nature 488:404-408 (2012), which is hereby incorporated by
reference in
its entirety. GSK-J4 has the structure of:
GSK-J4
41,
0
N
35
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Exemplary active derivatives of GSK4 that are also JMJD3 demthylase inhibitors
include GSK-J1 and GSK-J3, which have the following structures:
GSK JI
N rill
OH
ry
N N
0
X N
GSK-J3
KIII)
N INI
OH
tY
N N
0
H
.---'.
In another embodiment, the JMJD3 demethylase inhibitor comprises a modified
GSK-
J1 small molecule as described by Hu et aL, "Design and Discovery of New
Pyrimidine
Coupled Nitrogen Aromatic Rings as Chelating Groups of JMJD3 Inhibitors,"
Bioorg.
Med. Chem. Lett. 26(3):721-725 (2016), which is hereby incorporated by
reference in
its entirety.
In certain embodiments, the present disclosure provides a compound of formula
RilaN
....-----
NC
or a pharmaceutically acceptable salt thereof, wherein
36
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
R1 is -R, halogen, -OR, -SR, -N(R')2, -CN, -NO2, -C(0)R, -CO2R, -
C(0)N(R1)2, -C(0)SR, -C(0)C(0)R, -C(0)CH2C(0)R, -C(S)N(R)2, -C(S)OR,
-S(0)R, -SO2R, -SO2N(R)2, -N(R,C(0)R, -N(R1C(0)N(R1)2, -N(R)S02R,
-N(R`)S02N(R1)2, -N(R)N(R)2, -N(R')C(=N(R`))N(R1)2, -C=NN(R1)2, -
C=NOR, -C(=N(IT))N(R)2, -0C(0)R, or -0C(0)N(R1)2, wherein R and R' are as
defined above and described herein. In some embodiments, R1 is hydrogen. In
some
embodiments, RI is optionally substituted C1-6 aliphatic. In certain
embodiments,
R1 is optionally substituted 01-6 alkyl, C2-6 alkenyl, or 02-6a1kyny1. In
certain
embodiments, R1 is optionally substituted C1-6 alkyl. In certain embodiments,
R1 is
methyl. In certain other embodiments, R1 is ethyl or tert-butyl. In some
embodiments,
R1 is -OR, -SR, or -N(R)2. In certain embodiments, R1 is -SR. In certain
embodiments, R1 is -NH2. In certain embodiments, R1 is -CN or -NO2. In some
embodiments, R1 is halogen. In certain embodiments, R1 is fluoro, chloro,
bromo, or
iodo. In certain embodiments, R1 is fluor . In some embodiments, R1 is -C(0)R,
-
CO2R, -C(0)SR, -C(0)N(R1)2, -C(0)C(0)R, or -C(0)CH2C(0)R. In certain
embodiments, R1 is -C(S)OR or -C(S)N(R1)2. In other embodiments, R1 is -
S(0)R, -502R, or -502N(R')2. In some embodiments, R1 is -N(R')C(0)R, -
N(R)C(0)N(R)2, -N(R)S02R, -N(R')S02N(R12, -N(R1N(R1)2, or -
N(R)C(=N(RTN(R1)2. In certain embodiments, R1 is -N(R`)N(R1)2. In some
embodiments, R1 is -C=NN(R')2, -C=NOR, -C(=N(RI))N(R1)2, -0C(0)R, or -
0C(0)N(R)2;
Ring A is
37
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
N
)\).
X R3
N R3
R5
CY--R5
R2
N N
N R3,
N R3,
0
0--R5
)4-N.õõ,R2,
, or
N
R3
R6
R6
Rs
0
Ri,
R3
R6
wherein X, R2, R2', R3, R5, and R6 are as defined above and described herein.
Thus,
in certain embodiments, a compound of the disclosure is of one of the
following
formulae:
38
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
o-A5
R2
cX R3
N R3
NC NC
0
N
'
it'iy.1 N 11 R3 RI
N
R3
NC NC
0
2'
or
R1 ____________________________________________ ciy, RI N N
14
R3
NC NC
R6
0--Rs
RI ____________________________________________________________ / N
R3
NC
Re
wherein R1, R2, R2', R3, R5, R6, and X are as defined above and described
herein.
As defined generally above, R2 is ¨R, halogen, ¨OR, ¨SR, ¨N(R')2, ¨CN, ¨NO2,
¨C(0)R, ¨CO2R, ¨C(0)N(R1)2, ¨C(0)SR, ¨C(0)C(0)R, ¨C(0)CH2C(0)R, ¨
5 C(S)N(R')2, ¨C(S)OR, ¨S(0)R, ¨802R, ¨SO2N(R')2, ¨N(R')C(0)R, ¨
N(R)C(0)N(R)2, ¨N(RF)S02R, ¨N(R')S02N(R12, ¨N(R`)N(R`)2, ¨
N(T)C(=N(R))N(R1)2, ¨C=NN(R')2, ¨C=NOR, ¨C(=N(IT))N(R12, ¨0C(0)R, or
¨0C(0)N(IT)2, wherein R and R' are as defined above and described herein. In
some
embodiments, R2 is hydrogen. In some embodiments, R2 is optionally substituted
C1-
6 aliphatic. In certain embodiments, R2 is optionally substituted C1-6 alkyl,
C2-
6 alkenyl, or C2-6 alkynyl. In certain embodiments, R2 is optionally
substituted C1-
6 alkyl. In certain embodiments, R2 is ethyl. In certain other embodiments, R2
is
methyl, propyl, isopropyl, butyl, or isobutyl. In some embodiments, R2 is C1-6
alkyl
39
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
substituted with an -OH or -0C1-6alkyl group. In certain embodiments, R2 is -
CH2CH2OH or -CH2CH2OCH3. In some embodiments, R2 is cycloalkyl. In certain
embodiments, R2 is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In
some
embodiments, R2 is optionally substituted C2-6 alkenyl. In certain
embodiments, R2 is
allyl. In some embodiments, R2 is optionally substituted C2-6 alkynyl. In
certain
embodiments, R2 is 2-propynyl. In some embodiments, R2 is optionally
substituted
benzyl. In certain embodiments, R2 is unsubstituted benzyl. In certain other
embodiments, R2 is substituted benzyl. In some embodiments, R2 is C1-6 alkyl
substituted with an ester group. In certain embodiments, R2 is -CH2CO2C1-
6alkyl
or -CH2CO2aryl. In certain embodiments, R2 is -CH2CO2CH2CH3. In some
embodiments, R2 is -OR, -SR, or -N(IT)2. In certain embodiments, R2 is -CN or
-NO2. In some embodiments? R2is halogen. In certain embodiments, R2 is fluoro,
chloro, bromo, or iodo. In some embodiments, R2 is -C(0)R, -CO2R, -C(0)SR,
-C(0)N(IT)2, -C(0)C(0)R, or -C(0)CH2C(0)R. In certain embodiments, R2 is -
C(S)OR or -C(S)N(IT)2. In other embodiments, R2 is -S(0)R, -SO2R, or -
SO2N(IT)2. In some embodiments, R2 is -N(T)C(0)R, -N(T)C(0)N(R1)2, -
N(T)SO2R, -N(T)802N(T)2, -N(IT)N(IT)2, or -N(IT)C(=NOTDN(T)2. In some
embodiments, R2 is -C=NN(IT)2, -C=NOR, -C(=N(R))N(T)2, -0C(0)R, or -
OC(0)N(R')2.
As defined generally above, R2' is -R, -OR, -SR, -N(IT)2, -C(0)R, -CO2R, -
C(0)N(T)2, -C(0)SR, -C(0)C(0)R, -C(0)CH2C(0)R, -C(S)N(IT)2, -C(S)OR,
-S(0)R, -SO2R, -SO2N(IT)2, -N(IT)C(0)R, -N(IT)C(0)N(IT)2, -N(T)SO2R,
-N(T)S02N(T)2, -N(R')N(R')2, -N(R')C(=N(R`))N(IT)2, -C=NN(T)2, -
C=NOR, -C(=N(R`))N(IT)2, -0C(0)R, or -0C(0)N(T)2, wherein R and IT are as
defined above and described herein. In some embodiments, R2' is hydrogen. In
some
embodiments, R2' is optionally substituted C1-6 aliphatic. In certain
embodiments,
R2' is optionally substituted CI-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl. In
certain
embodiments, R2' is optionally substituted C1-6 alkyl. In certain embodiments,
R2' is
ethyl. In certain other embodiments, R2' is methyl, propyl, isopropyl, butyl,
or isobutyl.
In some embodiments, RZ is C1-6 alkyl substituted with an -OH or -0C1-6alkyl
group. In certain embodiments, R2' is -CH2GH2OH or -CH2CH2OCH3. In some
embodiments, R2' is cycloalkyl. In certain embodiments, R2' is cyclopropyl,
cyclobutyl,
cyclopentyl, or cyclohexyl. In some embodiments, R2' is optionally substituted
C1-
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
6 alkenyl. In certain embodiments, R2' is allyl. In some embodiments, R2'is
optionally
substituted C1-6 alkynyl. In certain embodiments, R2 is 2-propynyl. In some
embodiments, R2' is optionally substituted benzyl. In certain embodiments, R2'
is
unsubstituted benzyl. In certain other embodiments, R2' is substituted benzyl.
In some
embodiments, R2' is C1-6 alkyl substituted with an ester group. In certain
embodiments, R2'is -CH2CO2C1-6a1ky1 or -CH2CO2aryl. In certain embodiments,
R2 is -CH2CO2CH2CH3. In some embodiments, R2' is -OR, -SR, or -N(R52. In
some embodiments, R2' is -C(0)R, -CO2R, -C(0)SR, -C(0)N(R12, -
C(0)C(0)R, or -C(0)CH2C(0)R. In certain embodiments, R2'is -C(S)OR or -
C(S)N(R')2. In other embodiments, R2' is-S(0)R, -SO2R, or -SO2N(R`)2. In some
embodiments, R2' is -N(R)C(0)R, -N(R1C(0)N(R1)2, -N(R)SO2R, -
N(R)S02N(R1)2, -N(R)N(R1)2, or -N(R)C(=N(R))N(R1)2. In some embodiments,
R2' is -CsIN(R')2, -CsJOR, -C(il(R`))N(R1)2, -0C(0)R, or -0C(0)N(R1)2.
As defined generally above, R3 is -R, halogen, -OR, -SR, -N(R1)2, -CN, -NO2,
-C(0)R, -CO2R, -C(0)N(R)2, -C(0)C(0)R, -C(0)CH2C(0)R, -3(0)R, -
SO2R, -SO2N(R12, -N(R')C(0)R, -N(R)C(0)N(R)2, -N(IT)SO2R, -
N(R)S02N(R1)2, -N(R`)N(R')2, -C=NN(R1)2, -C=NOR, -0C(0)R, or -
0C(0)N(R1)2, wherein R and R' are as defined above and described herein. In
some
embodiments, R3 is hydrogen_ In some embodiments, R3 is optionally substituted
C1-
6aliphatic. In certain embodiments, R3 is optionally substituted C1-6 alkyl,
C2-
6 alkenyl, or C2-6 alkynyl. In certain embodiments, R3 is optionally
substituted C1-
6 alkyl. In certain embodiments, R3 is methyl. In certain other embodiments,
R3 is
ethyl, propyl, isopropyl, butyl, or isobutyl. In certain embodiments, R3 is -
CF3. In
some embodiments, R3 is C1-6 alkyl substituted with an -OH or -0C1-6alkyl
group.
In certain embodiments, R3 is -CH2OH, -CH2CH2OH, -CH2CH2CH2OH, -
CH2OCH2CH3, -CH2OCH3, -CH2CH2CH2OCH3, -CH(OH)CH3, or -
CH2CH2OCH3. In some embodiments, R3 is C1-6 alkyl substituted with an -NHC1-
6a1ky1 or -N(C1-6a1ky1)2 group_ In certain embodiments, R3 is -CH2NHC1-6a1ky1.
In certain embodiments, R3 is -CH2NHCH3. In some embodiments, R3 is C1-6 alkyl
substituted with an aryl, heteroaryl, carbocyclyl, or heterocyclyl ring. In
some
embodiments, R3 is optionally substituted benzyl. In certain embodiments, R3
is
unsubstituted benzyl. In certain other embodiments, R3 is substituted benzyl_
In
certain embodiments, R3 is -C(R0)2Ph. In certain embodiments, R3 is -C(R0)2Ph,
41
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
wherein Ro is hydrogen or methyl. In certain embodiments, R3 is
trifluoromethylbenzyl. In certain embodiments, R3 is ¨C(R0)2(heteroary1). In
certain
embodiments, R3 is ¨C(R0)2(heteroary1), wherein the heteroaryl is pyridyl,
pyrimidyl,
pyrazinyl, pyridazinyl, triazinyl, pyridinonyl, pyrrolyl, pyrazolyl,
imidazolyl, triazolyl,
tetrazolyl, thienyl, furanyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl,
isoxazolyl, or
oxadiazolyl. In certain embodiments, R3 is ¨CH2(heteroary1), wherein the
heteroaryl
is pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazinyl, pyrrolyl, pyrazolyl,
imidazolyl,
triazolyl, tetrazolyl, thienyl, furanyl, thiazolyl, isothiazolyl,
thiadiazolyl, oxazolyl,
isoxazolyl, or oxadiazolyl. In certain embodiments, R3 is
¨C(R0)2(carbocycly1). In
certain embodiments, R3 is ¨C(R0)2(carbocycly1), wherein the carbocyclyl is
cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl. In certain
embodiments, R3 is ¨CH2(carbocycly1), wherein the carbocyclyl is cyclopropyl,
cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl. In certain embodiments,
R3 is ¨
C(R0)2(heterocycly1). In certain embodiments, R3 is ¨C(R0)2(heterocycly1),
wherein
the heterocyclyl is tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl,
pyrrolidinyl,
pyrrolidonyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl,
tetrahydroisoquinolinyl,
decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl,
diazepinyl,
oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl. In certain
embodiments, R3 is
¨CH2(heterocycly1), wherein the heterocyclyl is tetrahydrofuranyl,
tetrahydrothienyl,
tetrahydropyranyl, pyrrolidinyl, pyrrolidonyl, piperidinyl, pyrrolinyl,
tetrahydroquinolinyl,
tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl,
dioxanyl,
dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and
quinuclidinyl. In some
embodiments, R3 is optionally substituted C2-6 alkenyl. In certain
embodiments, R3 is
allyl. In some embodiments, R3 is optionally substituted C2-6 alkynyl. In
certain
embodiments, R3 is propargyl. In some embodiments, R3 is an optionally
substituted
aryl or heteroaryl group. In certain embodiments, R3 is phenyl. In certain
embodiments, R3 is substituted phenyl. In certain embodiments, R3 is toluyl.
In certain
other embodiments, R3is a 5-6 membered heteroaryl ring having 1-3 heteroatoms
selected from nitrogen, oxygen, and sulfur. In certain embodiments, R3 is
pyridyl,
pyrimidyl, pyrazinyl, pyridazinyl, triazinyl, pyrrolyl, pyrazolyl, imidazolyl,
triazolyl,
tetrazolyl, thienyl, furanyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl,
isoxazolyl, or
oxadiazolyl. In some embodiments, R3 is ¨OR, ¨SR, or ¨N(R)2. In some
embodiments, R3 is halogen. In certain embodiments, R3 is fluoro, chloro,
bromo, or
42
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
iodo. In some embodiments, R3 is ¨C(0)R, ¨CO2R, ¨C(0)N(R1)2, ¨C(0)SR, ¨
C(0)C(0)R, or ¨C(0)CH2C(0)R. In certain embodiments, R3 is optionally
substituted ¨CO2C1-6alkyl. In certain embodiments, R3 is ¨0O2Et or ¨0O2Bn. In
certain embodiments, R3 is ¨CONHC1-6alkyl. In certain embodiments, R3 is ¨
CONHCH3 or ¨CONHCH2CH3. In certain embodiments, R3 is ¨C(S)OR or ¨
C(S)N(R')2. In other embodiments, R3 is ¨S(0)R, ¨SO2R, or ¨SO2N(R')2. In some
embodiments, R3 is ¨N(R)C(0)R, ¨N(R1C(0)N(R1)2, ¨N(R)S02R, ¨
N(R)S02N(RI)2, ¨N(R)N(R1)2, or ¨N(R')C(=N(R))N(R')2. In some embodiments,
R3 is ¨CAN(R`)2, ¨C=NOR, ¨C(=N(R))N(R1)2, ¨0C(0)R, or ¨0C(0)N(R)2.
In some embodiments, R2 and R3 are taken together with their intervening atoms
to
form an optionally substituted 5-7 membered partially unsaturated or aromatic
fused
ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and
sulfur.
In certain embodiments, R2 and R3 are taken together with their intervening
atoms to
form a 5-membered fused ring. In certain embodiments, R2 and R3 are taken
together
with their intervening atoms to form a fused cyclopentene ring. In certain
embodiments, R2 and R3 are taken together with their intervening atoms to form
a 6-
membered fused ring. In certain embodiments, R2 and R3 are taken together with
their intervening atoms to form a fused cyclohexene ring. In certain
embodiments,
R2 and R3 are taken together with their intervening atoms to form a fused
benzene
ring. In certain embodiments, R2 and R3 are taken together with their
intervening
atoms to form a 5-7 membered partially unsaturated fused ring having 1-4
heteroatoms independently selected from nitrogen, oxygen, and sulfur. In
certain
embodiments, R2 and R3 are taken together with their intervening atoms to form
a 5-
7 membered aromatic fused ring having 1-4 heteroatoms independently selected
from
nitrogen, oxygen, and sulfur.
In some embodiments, R2' and R3 are taken together with their intervening
atoms to
form an optionally substituted 5-7 membered partially unsaturated or aromatic
fused
ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and
sulfur.
In certain embodiments, R2' and R3 are taken together with their intervening
atoms to
form a 5-membered fused ring. In certain embodiments, R2' and R3are taken
together
with their intervening atoms to form a 6-membered fused ring. In certain
embodiments,
R2' and R3 are taken together with their intervening atoms to form a fused
pyridine
ring. In certain embodiments, R2' and R3 are taken together with their
intervening
43
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
atoms to form a 5-7 membered partially unsaturated fused ring having 1-4
heteroatoms independently selected from nitrogen, oxygen, and sulfur. In
certain
embodiments, R2' and R3 are taken together with their intervening atoms to
form a 5-
7 membered aromatic fused ring having 1-4 heteroatoms independently selected
from
nitrogen, oxygen, and sulfur.
As defined generally above, X is ¨N(R4)¨, ¨0¨, or ¨S¨, wherein R4 is as
defined
above and described herein. In certain embodiments, X is ¨0¨ or ¨S¨. In some
embodiments, X is ¨N(R4)¨. In certain embodiments, X is ¨NH¨. In certain
embodiments, X is ¨N(CH3)¨.
As defined generally above, R4 is ¨R, ¨C(0)R, ¨CO2R, or ¨S(0)2R, or R4 and
R3 are taken together with their intervening atoms to form an optionally
substituted 5-
7 membered saturated, partially unsaturated, or aromatic fused ring having 1-4
heteroatoms independently selected from nitrogen, oxygen, and sulfur. In
certain
embodiments, R4 is hydrogen. In some embodiments, R4 is optionally substituted
C1-
6 alkyl. In certain embodiments, R4 is optionally substituted C1-3 alkyl. In
certain
embodiments, R4 is methyl. In certain embodiments, R4 is substituted C1-6
alkyl. In
certain embodiments, R4 is benzyl. In certain embodiments, R4 is ¨
CH2CH2N(CH3)2. In some embodiments, R4 is awl or heteroaryl. In certain
embodiments, R4 is phenyl. In some embodiments, R4 is ¨C(0)R, ¨CO2R, or ¨
S(0)2R.
In some embodiments, R4 and R3 are taken together with their intervening atoms
to
form an optionally substituted 5-7 membered saturated, partially unsaturated,
or
aromatic fused ring having 1-4 heteroatoms independently selected from
nitrogen,
oxygen, and sulfur. In certain embodiments, R4 and R3 are taken together with
their
intervening atoms to form a 5-membered fused ring. In certain embodiments, R4
and
R3 are taken together with their intervening atoms to form a fused pyrrolidine
ring_ In
certain embodiments, R4 and R3 are taken together with their intervening atoms
to
form a 6-membered fused ring. In certain embodiments, R4 and R3 are taken
together
with their intervening atoms to form a fused piperidine ring. In certain
embodiments,
R4 and R3 are taken together with their intervening atoms to form a 5-7
membered
partially unsaturated fused ring having 1-4 heteroatoms independently selected
from
nitrogen, oxygen, and sulfur. In certain embodiments, R4 and R3 are taken
together
44
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
with their intervening atoms to form a 5-7 membered aromatic fused ring having
1-4
heteroatoms independently selected from nitrogen, oxygen, and sulfur.
As defined generally above, R5 is R, ¨C(0)R, ¨CO2R, ¨C(0)N(R')2, ¨C(0)C(0)R,
or ¨C(0)CH2C(0)R, or R5 and R2 are taken together with their intervening atoms
to
form an optionally substituted 5-7 membered partially unsaturated or aromatic
fused
ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and
sulfur.
In some embodiments, R5 is hydrogen. In some embodiments, R5 is optionally
substituted C1-6 alkyl. In certain embodiments, R5 is methyl. In certain
embodiments,
R5 is substituted C1-6 alkyl. In certain embodiments, R5 is C1-6 alkyl
substituted with
an ¨OH or ¨0C1-6 alkyl group. In certain embodiments, R5 is ¨CH2CH2OCH3. In
some embodiments, R4 is ¨C(0)R, ¨CO2R, ¨C(0)N(R1)2, ¨C(0)C(0)R, or ¨
C(0)CH2C(0)R.
As defined generally above, R6 is ¨R, halogen, ¨OR, ¨SR, ¨N(R')2, ¨CN, ¨NO2,
¨C(0)R, ¨CO2R, ¨C(0)N(R)2, ¨C(0)SR, ¨C(0)C(0)R, ¨C(0)CH2C(0)R, ¨
C(S)N(R')2, ¨C(S)OR, ¨S(0)R, ¨SO2R, ¨SO2N(R1)2, ¨N(R)C(0)R, ¨
N(R)C(0)N(R)2, ¨N(RISO2R, ¨N(R1)S02N(R)2, ¨N(R`)N(R1)2, ¨
N(RDC(=N(R))N(R')2, ¨CNN(R1)2, ¨C=NOR, ¨C(=N(R1))N(RI)2, ¨0C(0)R, or
¨0C(0)N(R1)2, wherein R and R' are as defined above and described herein. In
some
embodiments, R6 is hydrogen_ In some embodiments, R6 is optionally substituted
Cl -
6 aliphatic. In certain embodiments, R6 is optionally substituted C1-6 alkyl,
C2-
6 alkenyl, or C2-6alkynyl. In certain embodiments, R6 is optionally
substituted Cl-
6 alkyl. In certain embodiments, R6 is ethyl. In certain other embodiments, R6
is
methyl, propyl, isopropyl, butyl, or isobutyl. In some embodiments, R6 is C1-6
alkyl
substituted with an ¨OH or ¨0C1-6alkyl group. In certain embodiments, R6 is ¨
CH2CH2OH or ¨CH2CH2OCH3. In some embodiments, R6 is cycloalkyl. In certain
embodiments, R6 is cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In
some
embodiments, R6 is optionally substituted C2-6 alkenyl. In certain
embodiments, R6 is
allyl. In some embodiments, R6 is optionally substituted C2-6 alkynyl. In
certain
embodiments, R6 is 2-propynyl. In some embodiments, R6 is optionally
substituted
benzyl. In certain embodiments, R6 is unsubstituted benzyl. In certain other
embodiments, R6 is substituted benzyl. In some embodiments, R6 is C1-6 alkyl
substituted with an ester group. In certain embodiments, R6is ¨CH2CO2C1-6alkyl
or
¨CH2CO2aryl. In certain embodiments, R6 is ¨CH2CO2CH2CH3. In some
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
embodiments, R6 is ¨OR, ¨SR, or ¨N(R52. In certain embodiments, R6 is ¨CN or
¨NO2. In some embodiments, R6is halogen. In certain embodiments, R6 is fluoro,
chloro, bromo, or iodo. In some embodiments, R6 is ¨C(0)R, ¨CO2R, ¨C(0)SR,
¨C(0)N(R12, ¨C(0)C(0)R, or ¨C(0)CH2C(0)R. In certain embodiments, R6 is ¨
C(S)OR or ¨C(S)N(R')2. In other embodiments, R6 is ¨S(0)R, ¨SO2R, or ¨
SO2N(R12. In some embodiments, R6 is ¨N(R')C(0)R, ¨N(R)C(0)N(R1)2, ¨
N(R)S02R, ¨N(R')S02N(R1)2, ¨N(R`)N(R)2, or ¨N(R1C(=N(R))N(R)2. In some
embodiments, R6 is ¨C=NN(R')2, ¨C=NOR, ¨C(=N(R'))N(R)2, ¨0C(0)R, or ¨
0C(0)N(R1)2.
In some embodiments, R6 and R3 are taken together with their intervening atoms
to
form an optionally substituted 5-7 membered partially unsaturated or aromatic
fused
ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, and
sulfur.
In certain embodiments, R6 and R3 are taken together with their intervening
atoms to
form a 5-membered fused ring. In certain embodiments, R6 and R3 are taken
together
with their intervening atoms to form a fused cyclopentene ring. In certain
embodiments, R6 and R3 are taken together with their intervening atoms to form
a 6-
membered fused ring. In certain embodiments, R6 and R3 are taken together with
their intervening atoms to form a fused cyclohexene ring. In certain
embodiments,
R6 and R3 are taken together with their intervening atoms to form a fused
benzene
ring. In certain embodiments, R6 and R3 are taken together with their
intervening
atoms to form a 5-7 membered partially unsaturated fused ring having 1-4
heteroatoms independently selected from nitrogen, oxygen, and sulfur. In
certain
embodiments, R6 and R3 are taken together with their intervening atoms to form
a 5-
7 membered aromatic fused ring having 1-4 heteroatoms independently selected
from
nitrogen, oxygen, and sulfur.
As defined generally above, each R is independently hydrogen or an optionally
substituted group selected from C1-6 aliphatic, phenyl, a 3-7 membered
saturated or
partially unsaturated carbocyclic ring, an 8-10 membered bicyclic saturated,
partially
unsaturated or aryl ring, a 5-6 membered monocyclic heteroaryl ring having 1-3
heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 4-7
membered
saturated or partially unsaturated heterocyclic ring having 1-2 heteroatoms
independently selected from nitrogen, oxygen, or sulfur, a 7-10 membered
bicyclic
saturated or partially unsaturated heterocyclic ring having 1-4 heteroatoms
46
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered
bicyclic
heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen,
oxygen,
or sulfur. In certain embodiments, R is hydrogen. In some embodiments, R is
optionally
substituted C1-6 alkyl, C2-6 alkenyl, or C2-6 alkynyl. In certain embodiments,
R is
optionally substituted C1-6 alkyl. In certain embodiments, R is unsubstituted
C1-
6 alkyl. In certain embodiments, R is substituted C1-6 alkyl. In certain
embodiments,
R is methyl, ethyl, propyl, butyl, isopropyl, isobutyl, allyl, or benzyl.
In some embodiments, R is a 3-7 membered saturated or partially unsaturated
carbocyclic ring. In certain embodiments, R is a 3-4 membered saturated
carbocyclic
ring. In other embodiments, R is a 5-7 membered saturated or partially
unsaturated
carbocyclic ring. In certain embodiments, R is cyclopropyl, cyclobutyl,
cyclopentyl,
cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, or cycloheptenyl.
In some embodiments, R is a 4-7 membered saturated or partially unsaturated
heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen,
oxygen, or sulfur. In certain embodiments, R is a 4-7 membered saturated
heterocyclic
ring. In other embodiments, R is a 5-7 membered partially unsaturated
heterocyclic
ring. In certain embodiments, R is tetrahydrofuranyl, tetrahydrothienyl,
tetrahydropyranyl, pyrrolidinyl, pyrrolidonyl, piperidinyl, pyrrolinyl,
oxazolidinyl,
piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, or
morpholinyl.
In some embodiments, R is an 8-10 membered bicyclic saturated or partially
unsaturated carbocylic ring or a 7-10 membered bicyclic saturated or partially
unsaturated heterocyclic ring having 1-4 heteroatoms independently selected
from
nitrogen, oxygen, or sulfur. In certain embodiments, R is decahydronaphthyl,
tetrahydronaphthyl, or decalin. In certain other embodiments, R is
tetrahydroquinolinyl,
tetrahydroisoquinolinyl, or decahydroquinolinyl. In some embodiments, R is a
heterocyclyl ring is fused to an aryl or heteroaryl ring. In certain
embodiments, R is
indolinyl, 3H-indolyl, chromanyl, phenanthridinyl, 2-
azabicyclo[2.2.1]heptanyl,
octahydroindolyl, or tetrahydroquinolinyl.
In some embodiments, R is phenyl or a 5-6 membered heteroaryl ring having 1-3
heteroatoms independently selected from nitrogen, oxygen, or sulfur. In
certain
embodiments, R is phenyl. In certain other embodiments, R is a 5-membered
heteroaryl ring having 1-3 heteroatoms selected from nitrogen, oxygen, or
sulfur. In
47
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
yet other embodiments, R is a 6-membered heteroaryl ring having 1-3 nitrogens.
In
certain embodiments, R is phenyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl,
or triazinyl.
In certain other embodiments, R is pynrolyl, pyrazolyl, imidazolyl, triazolyl,
tetrazolyl,
thienyl, furanyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl, isoxazolyl,
or oxadiazolyl.
In some embodiments, R is bicyclic aromatic ring. In certain embodiments, R is
naphthyl. In other embodiments, R is an 8-10 membered bicyclic heteroaryl ring
having
1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In
certain
embodiments, R is quinolinyl, quinoxalinyl, quinazolinyl, pyridopyrazinyl, or
pyridopyrimidyl. In certain other embodiments, R is indolyl, benzimidazolyl,
benzothiazolyl, benzofuranyl, benzotriazolyl, benzoxazolyl, benzothiophenyl,
indazolyl, innidazopyridyl, innidazopyrirnidyl, innidazopyrazinyl,
imidazopyridazinyl,
pyrazolopyridyl, pyrazolopyrim idyl,
pyrazolopyrazinyl, pyrazolopyridazinyl,
pyrrolothiazolyl, im idazo-thiazolyl,
thiazolopyridyl, thiazolopyrimidyl,
thiazolopypyrazinyl, thiazolopyridazinyl,
oxazolopyridyl, oxazolopyrimidyl,
oxazolopyrazinyl, or oxazolopyridazinyl.
As defined generally above, each R' is independently ¨R, ¨C(0)R, ¨CO2R, or two
R' on the same nitrogen are taken together with the intervening nitrogen to
form a 4-7
membered heterocyclic ring having 1-2 heteroatoms independently selected from
nitrogen, oxygen, and sulfur. In certain embodiments, R' is R as defined and
described
above. In certain embodiments, R` is ¨C(0)R or ¨CO2R. In some embodiments, two
R' on the same nitrogen are taken together with their intervening atoms to
form a 4-7
membered heterocyclic ring having 1-2 heteroatoms independently selected from
nitrogen, oxygen, and sulfur. In certain embodiments, two R' on the same
nitrogen are
taken together with their intervening atoms to form an azetidine, pyrrolidine,
piperidine,
morpholine, piperazine, homopiperidine, or homopiperazine ring.
According to one aspect, a provided compound is of formula:
48
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
0
N- TN
c.....e.,,,,,
R1 ___________________________________________________
1
N
R3
NC I
R4
or a pharmaceutically acceptable salt thereof, wherein RI, R2, R3, and R4 are
as
defined and described herein. In certain embodiments, a compound of formula II
has
one of the following formulae:
II-a
N,_ N
Itt¨ /9:15:). .
NC I'
R4
11-1)
0
RI )50
N
NC I
R4 or
II-c
0
N-...Th
RI
NC I'
R4
According to another aspect, a provided compound is of formula III:
III
R2
RI ¨ciN )05C
0 R3
NC
49
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, and R4 are
as
defined and described herein. In certain embodiments, a compound of formula II
has
one of the following formulae:
III-a
N
R1 c____LTA113 ,
0
NC
111-11)
0
R1 __________________________________________________ ,,OF
0
NC
111-a
0
RI cliiiN 1
0
NC
According to another aspect, a provided compound is of formula:
R5
0"---
...õ1,..R2
N--,N
N
R3,
NC
or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, and R5 are
as
defined and described herein. In certain embodiments, R5 is optionally
substituted C1-
6 aliphatic. In certain embodiments, R5 is methyl. In some embodiments, R5is
optionally substituted C1-6 alkyl. In certain embodiments, R5 is substituted
C1-6 alkyl.
In certain embodiments, R5 is C1-6a1ky1 substituted with ¨OH or ¨0C1-6alkyl.
In
certain embodiments, R5 is ¨CH2CH20Me.
According to another aspect, a provided compound is of formula:
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
o
X Rr,
N.._
,--
RIti
N
R3
NC
or a pharmaceutically acceptable salt thereof, wherein R1, R2', and R3 are as
defined
and described herein.
Exemplary compounds that can be used as anti-PESC agents are set forth in
Table 1
below.
TABLE 1
0
icar0,
1-1
NC H
0
iciljW
1-2
-- N----
II
NC
1-3
NC
N
14
NC
51
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
0
ciL
NC
0
cct)It 1-6
NC
0
I
1-7
NC
0
Nõ,
I
1-8
N
NC
1-9
NC
0
OH
N-õ
yj
NC
o ,N
N
N
I-li
cL I
NC
52
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
tc1)05,
1-12
NC
0
_______________________________________________________________________________
_______ 1)5 1-13
NC
0
1-14
NC
"INC1)1C
c1H 14 I
1-15
NC
1-16
NC
0
< I
1-17
'1µ1"
El
NC
/14-4)51 0N
1-18
NC
110
53
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
0
1-19
NC
4111
1-20
NC
0
IC__ 14
j& I 1-21
OH
NC
0
cX1 I 1-22
OH
NC
0
N
CI I 1-23
NC
0
I 1-24
NC
54
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
0
N.,
1-25
o----,---
N
ti
NC
0
0
Ci I
1-26
r-----Nnro-,
H
NC
0
0----'''''-'=-A-'''
1-27
N
NC
0
0
y_ I
1-28
N
H
NC
1-29
N
NC
C-- l'1)5C01
0
c_CC
1-30
N
NC
0
7 I
1-31
irc---
H
NC
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
0
NN I
1-32
N----"--./--.C)-,
FT
NC
0
< : I
1-33
H
NC
0
I 1-34
--- N
ET
NC
0
cILI I 1-35
N
H
NC
OH
0
N
1-36
NC
H
N
0
clIN I
1-37
N
NC
0
14, 1,1
c.õ,__L I OH 1-38
N
H
NC
56
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
C
1-39
c-1:IN )N
H
NC
0
N OH
N
H
NC
0
1-41
II 1S-----LI)N5CCF3
H
NC
0
cN
_____ I
1-42
N
NC
0
1
NC
0
0
Nõ
L I
N
1-44
H
NC
0
a
1-45
0,....õ.õ,......-
N
H
NC
57
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
0
N-,
c,"::( I
1-46
NC
0
N-õ
I
1-47
NC
0
I
1-48
NC
0
0
1-49
NC
0
0
1-50
NC
0
1-51
NC
0
(
1-52
NC
58
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
0
N
cL I
1-53
o
ri
IT
NC
0
-.,....0
N-__ ,-.=-)_CI
c_i_g --.
1-54
N
NC
0
N
cL I
1-55
N
H
NC
0
N,õ
cji I
1-56
N
H
NC
N-,
CF3
SlL1:1*HN
1-57
NC
N
crL)5CANCI
II
1-58
-___
NC
0,._,_õ.011
ci.,....,_ ',-
N-, _JD
1-59
N
NC
59
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
0
/14----N
1-60
N
H
NC
0
0
7--- N
Sc_j__ __.,JLY I
1-61
H
ri
H
NC
0
0
lck
.õ... N
1-62
c
N
NC
0
N----14A--"--
S_
,...,....,..cN
14
El
NC I
0
c________ I
1-64
N
1110
H
NC
__
HiN
1-65
ETY c)N
H
NC
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
0
N-_,
;c.:1 I
N
NC H
1-66
SF
F
F
0
N
cl, I
N
1-67
H
NC
I
N-...,,,...t.a.
0
7---- -L---'''''-
___IL,__1 I
N II
1-68
NC
N N
0
c__/ NN
I
N
1-69
II
NC _________________________________________________
IS
0
N
c____ I
N
1-70
H
NC
61
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
TABLE 1
0
N-, N
N
H
1-71
NC
1
..
N 0
H
0 e_..-
___I
14,,
, I
N
1-72
H
NC.
0
N
1-73
H
NC
We-'...
In certain embodiments, the present disclosure provides any compound depicted
in
Table 1, above, or a pharmaceutically acceptable salt thereof.
In other embodiments, the present disclosure provides a compound of formula:
0
R2
R 1 ¨c\IN
1
N
R3
NC I
R4
62
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
or a salt thereof, wherein:
R1 is H, C1-6a1ky1, trifluoromethyl, 3-6 membered carbocyclyl, 6 membered
aryl, 3-6
membered heterocyclyl, 5-6 membered heteroaryl, halo, -0Rf, -SRf, -N(Rf)2, -
CN, or -NO2, wherein said alkyl, carbocyclyl, aryl, heteroaryl, and
heterocyclyl are
optionally substituted with one or more groups independently selected from
oxo, halo,
C1-3alkoxy and C1-3a1ky1;
R2 and R3 are each independently H, C1-12alkyl, C2-12alkenyl, C2-12alkynyl,
carbocyclyl, aryl, heterocyclyl, heteroaryl, halo, -0Ra, -SRa, -N(Ra)2, -CN, -
NO2, -C(0)Ra, -CO2Ra, -C(0)N(Ra)2, -C(0)SRa, -C(0)C(0)Ra, -
C(0)CH2C(0)Ra, -C(S)N(Ra)2, -C(S)0Ra, -S(0)Ra, -SO2Ra, -SO2N(Ra)2,
-N(Ra)C(0)Ra, -N(Ra)C(0)N(Ra)2, -N(Ra)S02Ra, -N(Ra)S02N(Ra)2, -
N(Ra)N(Ra)2, -N(Ra)C(=N(Ra))N(Ra)2, -C(s1)N(Ra)2, -C=NORa, -
C(=N(Ra))N(Ra)2, -0C(0)Ra, or -0C(0)N(Ra)2, wherein each C1-12alkyl, C2-
12a1keny1, C2-12a1kyny1, carbocyclyl, aryl, heteroaryl, and heterocyclyl of R2
and R3 is
independently optionally substituted with one or more groups Rx; and wherein
R2 and
R3 are not each H; or R2 and R3 taken together with the atoms to which they
are
attached form a 4, 5, 6, 7, or 8 membered carbocyclyl or aryl, which
carbocyclyl or aryl
is optionally substituted with one or more groups Rx;
R4 is H, C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl, carbocyclyl, aryl,
heteroaryl, and
heterocyclyl, wherein each C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl,
carbocyclyl, aryl,
heteroaryl, and heterocyclyl is optionally substituted with one or more groups
independently selected from oxo, C1-12 alkyl, C1-12 haloalkyl, carbocyclyl,
aryl,
heterocyclyl, heteroaryl, halo, -CN, -NO2, -NRmRm, -ORm, -C(=0)0Rnn, and
-0C(=0)Rm; or R4 and R3 taken together with the atoms to which they are
attached
form a heterocyclyl;
each Ra is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6
alkynyl,
carbocyclyl, aryl, heteroaryl, and heterocyclyl, wherein each C1-6 alkyl, C2-6
alkenyl,
C2-6 alkynyl, carbocyclyl, aryl, heteroaryl, and heterocyclyl is optionally
substituted
with one or more groups Rx;
each Rf is independently selected from H, C1-3 alkyl, trifluoromethyl, 3-6
membered
carbocyclyl, 6 membered aryl, 3-6 membered heterocyclyl, and 5-6 membered
63
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
heteroaryl, or two Rf groups together with the nitrogen to which they are
attached form
a 3-6 membered heterocycle;
each Rg is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6
alkynyl, C3-
8 carbocyclyl, aryl, heteroaryl, and heterocyclyl, wherein each C1-6 alkyl, C2-
6 alkenyl,
C2-6 alkynyl, C3-8 carbocyclyl, aryl, heteroaryl, and heterocyclyl is
optionally
substituted with one or more groups Rx; or two Rg groups together with the
nitrogen
to which they are attached form a 3-6 membered heterocycle or a 5-6 membered
heteroaryl;
each Rm is independently selected from H, C1-6 alkyl, C2-6 alkenyl, C2-6
alkynyl, C1-
6 haloalkyl, carbocyclyl, C1-6 alkanoyl, phenyl, and benzyl, wherein any C1-6
alkyl,
C2-6 alkenyl, C2-6 alkynyl, C1-6 haloalkyl, carbocyclyl, C1-6 alkanoyl,
phenyl, or
benzyl is optionally substituted with one or more groups independently
selected from
halo, ¨CN, ¨NO2, ¨NRyRz, and ¨0Rw; or two Rm groups together with the
nitrogen to which they are attached form a 3-6 membered heterocycle;
each Rv is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl,
carbocyclyl, aryl, heteroaryl, and heterocyclyl, wherein each C1-6 alkyl, C2-6
alkenyl,
C2-6 alkynyl, carbocyclyl, aryl, heteroaryl, and heterocyclyl is optionally
substituted
with one or more groups independently selected from oxo, halo, amino,
hydroxyl, aryl,
carbocyclyl, and C1-C6 alkyl that is optionally substituted with one or more
groups
independently selected from oxo and halo; or two Rv are taken together with
the
nitrogen to which they are attached to form a heterocyclyl that is optionally
substituted
with one or more groups independently selected from oxo, halo and C1-3 alkyl
that is
optionally substituted with one or more groups independently selected from oxo
and
halo;
each Rw is independently selected from H, C1-4 alkyl, C1-4 alkanoyl, phenyl,
benzyl,
and phenethyl;
each Rx is independently selected from oxo, C1-6 alkyl, C2-6 alkenyl, C2-6
alkynyl,
carbocyclyl, aryl, heteroaryl, heterocycle, ¨F, ¨Cl, ¨Br, ¨I, ¨NO2, ¨N(Rv)2, ¨
CN, ¨C(0)¨N(Rv)2, ¨8(0)¨N(Rv)2, ¨8(0)2¨N(Rv)2, ¨0¨Rv, ¨8¨Rv, ¨0-
C(0)¨Rv, ¨0¨C(0)-0¨Rv, ¨C(0)¨Rv, ¨C(0)-0¨Rv, ¨3(0)¨Rv, ¨
S(0)2¨Rv, ¨0¨C(0)¨N(Rv)2, ¨N(Rv)¨C(0)-0Rv, ¨N(Rv)¨C(0)¨N(Rv)2,
64
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
-S(0 )2-N( Rv)2, -N( Rv)-C(0)-Rv, -N(Rv)-S(0)-Rv, -N(Rv)-S(0)2-Rv,
-N(Rv)-8(0)-N(Rv)2, and
-N(Rv)-S(0)2-N(Rv)2, wherein any C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl,
carbocyclyl, aryl, heteroaryl, and heterocycle is optionally substituted with
one or more
groups independently selected from Ftxa, oxo, halo, -NO2, -N(Rv)2, -CN, -
C(0)-N(Rv)2, -3(0)-N(Rv)2, -S(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-
C(0)-Rv, -C(0)-Rv, -C(0)-0-Rv, -3(0)-Rv, -S(0)2-Rv, -C(0)-
N(Rv)2, -8(0)2-N(Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-C(0)-0Rv, -N(Rv)-
S(0)-Rv, -N(Rv)-S(0)2-Rv, and C1-6 alkyl that is optionally substituted with
one
or more groups independently selected from oxo and halo;
each Ry and Rz is independently selected from H, C1-4 alkyl, C1-4 alkanoyl, C1-
4
alkoxycarbonyl, phenyl, benzyl, and phenethyl, or Ry and Rz together with the
nitrogen to which they are attached form a heterocyclyl;
each Rxa is independently selected from aryl, heteroaryl, heterocycle, and
carbocycle,
wherein any aryl, heteroaryl, heterocycle, and carbocycle is optionally
substituted with
one or more groups independently selected from C1-6 alkyl, C2-6 alkenyl, C2-6
alkynyl, -F, -Cl, -Br, -I, -NO2, -N(Rv)2, -CN, carbocycle, aryl, -C(0)-
N(Rv)2, -S(0)-N(Rv)2, -8(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-C(0)-Rv, -
0-C(0)-0-Rv, -C(0)-Rv, -C(0)-0-Rv, -S(0)-Rv, -S(0)2-Rv, -0-
C(0)-N(Rv)2, -N(Rv)-C(0)-0Rv, -N(Rv)-C(0)-N(Rv)2, -S(0)2-N(Rv)2,
-N(Rv)-C(0)-Rv, -N(Rv)-S(0)-Rv, -N(Rv)-8(0)2-Rv, and -N(Rv)-
S(0)-N(Rv)2, wherein any C1-6 alkyl, C2-6 alkenyl, and C2-6 alkynyl is
optionally
substituted with one or more groups independently selected from oxo, halo, -
NO2,
-N(Rv)2, -CN, -C(0)-N(Rv)2, -S(0)-N(Rv)2, -S(0)2-N(Rv)2, -0-Rv, -
S-Rv, -0-C(0)-Rv, -C(0)-Rv, -C(0)-0-Rv, -S(0)-Rv, -S(0)2-Rv,
-C(0)-N(Rv)2, -8(0)2-N(Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-S(0)-Rv, and
-N(Rv)-8(0)2-Rv.
In certain embodiments, R1 is H, C1-6 alkyl, trifluoromethyl, 3-6 membered
carbocyclyl, 6 membered aryl, 3-6 membered heterocyclyl, 5-6 membered
heteroaryl,
halo, -0Rf, -SRf, -N(Rf)2, -CN, or -NO2, wherein said alkyl, carbocyclyl,
aryl,
heteroaryl, and heterocyclyl are optionally substituted with one or more
groups
independently selected from oxo, halo, C1-3 alkoxy and C1-3 alkyl.
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In certain embodiments, R1 is H, methyl, or ethyl.
In certain embodiments, R1 is H.
In certain embodiments, R2 is H.
In certain embodiments, R2 is C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl,
carbocyclyl,
aryl, heterocyclyl, heteroaryl, halo, -0Ra, -SRa, -N(Ra)2, -CN, -NO2, -
C(0)Ra, -CO2Ra, -C(0)N(Ra)2, -C(0)SRa, -C(0)C(0)Ra, -C(0)CH2C(0)Ra,
-C(S)N(Ra)2, -C(S)0Ra, -S(0)Ra, -SO2Ra, -SO2N(Ra)2, -N(Ra)C(0)Ra, -
N(Ra)C(0)N(Ra)2, -N(Ra)S02Ra, -N(Ra)802N(Ra)2, -N(Ra)N(Ra)2, -
N(Ra)C(=N(Ra))N(Ra)2, -C(1)N(Ra)2, -C,=NORa, -C(sl(Ra))N(Ra)2, -
OC(0)Ra, or -0C(0)N(Ra)2, wherein each C1-12 alkyl, C2-12 alkenyl, C2-12
alkynyl, carbocyclyl, aryl, heteroaryl, and heterocyclyl of R2is independently
optionally
substituted with one or more groups Rx.
In certain embodiments, R2 and R3 taken together with the atoms to which they
are
attached form a 4, 5, 6, 7, or 8 membered carbocyclyl or aryl, which
carbocyclyl or aryl
is optionally substituted with one or more groups Rx.
In certain embodiments, R2 is H, C1-6 alkyl, C2-12 alkenyl, C2-12 alkynyl,
carbocyclyl,
aryl, heteroaryl, halo, -CN, -SRa, -N(Rv)2, and -CO2Ra, wherein any C1-6
alkyl,
carbocyclyl and aryl is optionally substituted with one or more groups
independently
selected from C1-3alkyl, carbocyclyl, halo, -CN, -N(Rv)-C(0)-Rv, and -0-Rv.
In certain embodiments, R2 is H, isopropyl, ethyl, tert-butyl, 2,2-
difluoroethyl,
cyclobutyl, 2-propyn-1-yl, bromo, chloro, 2-furyl, vinyl, phenyl, 2-
chlorophenylthio, 2-
fluoroethyl, 2-propenyl, 1-methylvinylcyclopropyl, 4-pyridyl, 2-buten-1-yl,
iodo, 1-
methyl-2-propyn-1-yl, 1-methylprop-1-yl, 1-(cyclopropyl)ethyl,
methoxycarbonyl, 2-
butynyl, 2-hydroxy-1-methylethyl, 4-
(methylcarbonylamino)butyl, 3-
(methylcarbonylamino)propyl, 4-aminobutyl, 1-methyl-2-propenyl, 1-
methylcyclobutyl,
propyl, 2-methoxyethyl, and 2-methylpropyl.
In certain embodiments, R3 is H.
In certain embodiments, R3 is C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl,
carbocyclyl,
aryl, heterocyclyl, heteroaryl, halo, -0Ra, -SRa, -N(Ra)2, -CN, -NO2, -
C(0)Ra, -CO2Ra, -C(0)N(Ra)2, -C(0)SRa, -C(0)C(0)Ra, -C(0)CH2C(0)Ra,
-C(S)N(Ra)2, -C(S)0Ra, -S(0)Ra, -SO2Ra, -SO2N(Ra)2, -N(Ra)C(0)Ra, -
66
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
N(Ra)C(0)N(Ra)2, -N(Ra)S02Ra, -N(Ra)S02N(Ra)2, -N(Ra)N(Ra)2, -
N(Ra)C(=N(Ra))N(Ra)2, -C(=N)N(Ra)2, -C=NORa, -C(=N(Ra))N(Ra)2, -
OC(0)Ra, or -0C(0)N(Ra)2, wherein each C1-12 alkyl, C2-12 alkenyl, C2-12
alkynyl, carbocyclyl, aryl, heteroaryl, and heterocyclyl of R3is independently
optionally
substituted with one or more groups Rx.
In certain embodiments, R3 is H, C1-12 alkyl, C2-12 alkenyl, C2-12 alkynyl,
aryl,
heterocyclyl, heteroaryl, halo, -0Ra, -N(Ra)2, -C(0)Ra, -CO2Ra, -
C(0)N(Ra)2, or -N(Ra)C(0)Ra, wherein each C1-12 alkyl, C2-12 alkenyl, C2-12
alkynyl, aryl, heteroaryl, and heterocyclyl of R3 is independently optionally
substituted
with one or more groups Rx.
In certain embodiments, R3 is H, methyl, chloro, bromo, carboxy, formyl,
aminocarbonyl, furan-3-yl, phenyl, benzyl, phenethyl, phenoxy, 1H-pyrazol-4-
yl, 1-
(cyclopropylmethyl)-1 H-pyrazol-4-yl, 1-(1-
methylcyclopropy1)-1H-pyrazol-4-yl, 5-
fluoro-1H-pyrazol-4-yl, 1-(2-phenylpropan-2-y1)-1H-pyrazol-4-yl, 1-(pyridin-3-
yI)-1H-
pyrazol-4-yl, 1-(pyridin-4-y1)-1H-pyrazol-4-yl, 1-(pyridin-2-y1)-1H-pyrazol-4-
yl, 141 -(N-
methylaminocarbony1)-1 ,1-dimethylmethy1]-1 H-pyrazol-4-yl, 5-fluoro-1-
isopropy1-1H-
pyrazol-4-yl, 1-(cyclopropyl-methyl)-1H-pyrazol-5-yl,
1-(cyclopropylmethyl)-1H-
pyrazol-3-yl, 1-(tetrahydro-2H-thio-pyran-4-y1)-1H-
pyrazol-4-yl, 1-(1 , 1-
dioxidotetrahydro-2H-thiopyran-4-y1)-1H-pyrazol-4-yl,
1-((6-(3-oxobut-1-en-1-
yl)pyridin-2-yl)methyl)-1H-pyrazol-4-yl, 3-iodophenyl, methyl-am inocarbonyl,
3-
methy1-1,2,4-oxad iazol-5-yl, 5-methyl-1,3,4-oxadiazol-2-yl, 1H-imidazol-2-yl,
N-
(benzoylmethyl)aminocarbonyl, 5-phenyloxazol-2-yl, 1-cyclohexyl-pyrazol-4-yl,
1-
isopropylpyrazol-4-yl, biphenyl-3-yl, 3-((4-
fluorophenyl)amino)phenyl, 3-(2-
oxopyrrolidin-1-yl)phenyl, 3-(methylcarbonylamino)-5-phenylphenyl, phenylam
ino,
piperidin-1-yl, methoxymethyl, ethoxymethyl, ethoxycarbonyl, 3-methoxypropyl,
benzyl-oxycarbonyl, trifluoromethyl, 3-furyl, ethylaminocarbonyl,
hydroxymethyl, 3-
hydroxypropyl, 2-hydroxyethyl, m ethylam inom ethyl , benzofuran-3-yl, 1-
pheny1-1H-
pyrazol-3-yl, 5-cyclopropylfuran-2-yl, 2-methylfuran-3-yl, 1-pheny1-1H-pyrazol-
4-yl, 1-
ethy1-1H-pyrazol-4-yl, 1-m ethy1-6-oxo-1 ,6-d
ihydropyridin-3-yl, furan-2-yl, 5-
phenylfuran-2-yl, 1-isopropy1-1H-pyrazol-4-yl, pyrimidin-5-yl, 5-methylpyridin-
3-yl, 1-
methy1-1H-pyrazol-3-yl, 4-phenylfuran-2-yl, 2-fluorophenyl, 4-cyanophenyl, 4-
methoxyphenyl, 4-(trifluoromethyl)phenyl, 4-fluorophenyl, 1-benzy1-1H-pyrazol-
4-yl, 5-
chloropyridin-3-yl, 5-fluoropyridin-3-yl, prop-1-en-2-yl, vinyl, 1-methyl-1H-
pyrazol-5-yl,
67
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
4-(hydroxymethyp-furan-2-yl, 3-cyanophenyl, 1 H-pyrazol-5-yl, 2,5-dihydrofuran-
3-yl,
thiophen-3-yl, thiophen-2-yl, 1-methyl-1H-pyrazol-4-yl, 5-methylfuran-2-yl, 5-
(hydroxym ethyl)furan-2-yl, 3-
(trifluoromethyl)phenyl, 3-m ethoxyphenyl, 3-
fluorophenyl, pyridin-3-yl, 1-(m ethylsulfony1)-IH-pyrazol-4-yl, 1-cyclopenty1-
1H-
pyrazol-4-yl, 1-(th rophen-3-ylmethyl)-1H-pyrazol-4-yl,
4-chloro-3-(morpholine-4-
carbonyl)phenyl, 3-chloro-4-(cyclopropyl-aminocarbonyl)phenyl, 1-(1-hydroxy-2-
methylpropan-2-y1)-1H-pyrazol-4-yl, 1-(3-methoxybenzy1)-1H-pyrazol-4-yl, 1-
(pyridin-
4-ylmethyl)-1H-pyrazol-4-yl, 1-(2-chloro-
benzy1)-1H-pyrazol-4-yl, 1-(3-
phenoxybenzy1)-1H-pyrazol-4-yl, 1-(4-phenoxybenzy1)-1H-pyrazol-4-yl, 1-
cyclohexyl-
1 H-pyrazol-4-yl, 1-(1-phenylethyl)-1H-pyrazol-4-yl, 1-cyclobuty1-1H-pyrazol-4-
yl, 1-
(sec-buty1)-1H-pyrazol-4-yl, 4-fluoro-3-
(pyrrolidine-1-carbonyl)phenyl, 1-
(cyclopropylsulfony1)-1H-pyrazol-3-yl, 1-(cyclopropanecarbony1)-1H-pyrazol-3-
yl, 1-
(2-cyclopropylethyl)-1H-pyrazol-4-yl, 1-([1,11-bipheny1]-3-ylmethyl)-1H-
pyrazol-4-yl, 1-
phenethy1-1H-pyrazol-4-yl, 1-(2-
methoxybenzy1)-1H-pyrazol-4-yl, 1-(4-
methoxybenzy1)-1H-pyrazol-4-yl, 1-(tert-buty1)-1H-pyrazol-4-yl, 3,4-
dimethylphenyl, 3-
chloro-4-ethoxyphenyl, 4-methoxy-3-methylphenyl, 2-methylbenzo[d]thiazol-5-yl,
1-
(2-phenoxybenzy1)-1H-pyrazol-4-yl, 1-(phenylsulfony1)-1H-pyrazol-4-yl, 1-
benzoy1-1H-
pyrazol-4-yl, 1-benzhydry1-1H-pyrazol-4-yl, 1-([1,1r-bipheny1]-2-ylmethyl)-1H-
pyrazol-
4-yl, 1-(cyclohexylmethyl)-1H-pyrazol-4-yl, 1-(pyridin-3-ylmethyl)-1H-pyrazol-
4-yl,
benzo-furan-2-yl, (E)-styryl, 5-ethylfuran-2-yl, 1-(2-methoxyethyl)-1H-pyrazol-
4-yl, 1-
(naphthalen-1-ylmethyl)-1 H-pyrazol-4-yl, 1-([1,1r-bipheny1]-4-ylmethyl)-1H-
pyrazol-4-
yl, 3-phenoxyphenyl, phenylethynyl, 3,4-dichlorophenyl, 3-chloro-4-
methoxyphenyl, 3-
methoxy-4-methylphenyl, 1-(thiazol-4-ylmethyl)-1H-pyrazol-4-yl, 1 H-indazol-5-
yl, 3,4-
dimethoxyphenyl, 4-methoxy-3,5-dimethylphenyl, 1-(oxetan-3-y1)-1H-pyrazol-4-
yl, 1-
(2-fluorobenzy1)-1H-pyrazol-4-yl, 1-(4-fluorobenzy1)-1H-pyrazol-4-yl, 1-
(methoxy-
carbonylmethyl)-1H-pyrazol-4-yl, 1-(2-
(dimethylam ino)ethyl)-1H-pyrazol-4-yl, 3-
cyano-4-methylphenyl, benzo[d][1,3]dioxo1-5-yl, 2,3-dihydrobenzofuran-5-yl, 1-
(3-
fluorobenzy1)-1H-pyrazol-4-yl, 1-(th iophen-2-
ylmethyl)-1H-pyrazol-4-yl, 142,2,2-
trifluoroethyl)-1 H-pyrazol-4-yl, 1-(3-chlorobenzy1)-
1H-pyrazol-4-yl, 1-isobuty1-1H-
pyrazol-4-yl, 1-(3,3,3-trifluoropropy1)-1H-pyrazol-4-yl, 1-(d ifluoromethyl)-1
H-pyrazol-4-
yl, 1-(2-cyanoethyl)-1H-pyrazol-4-yl, 4-cyclopropylfuran-2-yl, 1H-pyrrol-3-yl,
2,2-
d ifluorobenzo[d][1 ,3]clioxo1-5-yl, 3-fluoro-4-
(aminocarbonyl)phenyl, 3-fluoro-4-
(methylsulfonyl)phenyl, 3-chloro-4-
(tritluoromethoxy)phenyl, 5-fluoro-3-
(aminocarbonyl)phenyl, 3-(hydroxymethyl)-4-methoxyphenyl, 1-(methylsulfony1)-
1H-
68
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
pyrrol-3-yl, 1-methyl-1H-pyrrol-3-yl, 1H-indo1-2-yl, cyclopropylcarbonylamino,
benzoylamino, 3-bromophenyl, 3-(1-methylpyrazol-4-yl)phenyl, 3-(1-
isopropylpyrazol-
4-yl)phenyl, 4-phenylphenyl, 4-(4-
fluoroanilino)phenyl, 3-(tert-
butoxycarbonylamino)phenyl, 1-acety1-1,2,3,6-tetrahydropyridin-4-yl, 1-
propionyl-
1,2,3,6-tetrahydropyridin-4-yl, 1-acryloy1-1,2,3,6-tetrahydropyridin-4-yl, 1-
methyl-
1 ,2,3,6-tetrahydropyridin-4-yl, 1-02-methylthiazol-4-yOmethyl)-1H-pyrazol-4-
yl, 1-(2-
(acetyl-am ino)ethyl)-1H-pyrazol-4-yl,
3, 5-dichlorophenyl, 2-fluoro-4-
(methylsulfonyl)phenyl, 1-(tert-penty1)-1H-pyrazol-4-yl, 3-(2-
morpholinoethyl)phenyl,
3-(2-(dimethyl-amino)ethyl)-phenyl, 1-(1-
(thiazol-4-yl)ethyl)-1H-pyrazol-4-yl, 1-
(tetrahydro-2H-pyran-4-y1)-1H-pyrazol-4-yl, 3-methoxy-4-
(trifluoromethyl)phenyl, 3-
methoxycarbony1-4-chlorophenyl, 4-
(trifluoromethoxy)phenyl, 3-methy1-4-
(trifluorornethoxy)phenyl, 4-cyclopropy1-3-(trifluoro-methyl)phenyl, 2,2-
dimethy1-2,3-
dihydrobenzofuran-5-yl, 3,5-dim ethoxyphenyl, 3,4-difluorophenyl, 4-biphenyl,
3-
chloro-5-fluorophenyl, 3,5-bis(trifluoromethyl)phenyl, 3-fluoro-5-
methoxyphenyl, 3-
(am inocarbonyl)phenyl, 4-(cyclopropylmethoxy)phenyl,
2-fluoro-5-
(benzyloxycarbonyl)phenyl, 3-(1H-pyrazol-1-yl)phenyl, 1-(2-hydroxycyclo-
penty1)-1H-
pyrazol-4-yl, 3-(N-methylam inosulfonyl)phenyl, 4-(2-hydroxypropan-2-yI)-
phenyl, 2-
(trifluoromethyl)pyridin-4-yl, 6-phenoxypyridin-3-yl, 2-methoxypyridin-4-yl, 4-
methy1-2-
phenylthiazol-5-yl, 3-am ino-5-cyanophenyl,
1-(tetrahydrofuran-3-yl, 3-(N-
ethylaminocarbonyl)phenyl, 3-(aminocarbonylmethyl)phenyl, 6-phenylpyridin-3-
yl, 1-
(tetrahydro-2H-pyran-3-y1)-1H-pyrazol-4-yl, 1-(1-methoxypropan-2-y1)-1H-
pyrazol-4-
yl, 1-(2-ethoxyethyl)-1H-pyrazol-4-yl, 1-acety1-2,5-dihydro-1H-pyrrol-3-yl, 1-
acetyl-
1 ,2,5,6-tetrahydropyridin-3-yl, 1-propiony1-1,2,5,6-tetrahydropyridin-3-yl, 1-
propiony1-
2,5-dihydro-1H-pyrrol-3-yl, 1-((1S,38)-3-hydroxycyclobuty1)-1H-pyrazol-4-yl,
2,5-
dihydro-1H-pyrrol-3-yl, 1,2,5,6-tetrahydropyridin-
3-yl, 1-methy1-1,2,5,6-
tetrahydropyridin-3-yl, 1-acryloy1-1,2,5,6-
tetrahydropyridin-3-yl, 1-acryloy1-2,5-
dihydro-1H-pyrrol-3-yl, 4-chloro-3,5-dimethylphenyl, 4-cyano-3-methylphenyl, 1-
oxo-
2,3-dihydro-1H-inden-5-yl,
3,4-bis(trifluoromethyl)phenyl, 3-methy1-4-
(trifluoromethyl)phenyl, 1-(benzo[b]thiophen-7-ylmethyl)-1H-pyrazol-4-yl, 4-
fluoro-3-
(N-cyclohexylaminocarbonyl)phenyl, 4-m orpholino-
phenyl, 4-(4-(tert-
butoxycarbonyppiperazin-1-yl)phenyl,
3-chloro-5-methylphenyl, 3-
(methylsulfonyl)phenyl, 4-(methylsulfonylam ino)phenyl, 4-
(morpholinomethyl)phenyl,
3-morpholinophenyl, 1-(2-
(vinylcarbonylamino)ethyl)-1H-pyrazol-4-yl, 1-(2-
am inoethyl)-1H-pyrazol-4-yl, 3-cyclopropy1-4-methylphenyl, 3-ethoxyphenyl, 3-
69
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
(hydroxymethyl)phenyl, 1-(2-(tert-butoxycarbonylamino)ethyl)-1H-pyrazol-4-yl,
3-
phenethoxyphenyl, 1,2, 3,6-tetrahydropyridin-4-yl, 1-(2-
(vinylsulfonylamino)ethyl)-1H-
pyrazol-4-yl, 4-(phenylamino)-phenyl, 3-methyl-1H-pyrazol-4-yl, 4-
(benzyloxy)phenyl,
3,5-d ifluorophenyl, 3-fluoro-5-trifluoromethylphenyl, 3-
(ethylsulfonyl)phenyl, 3-
(trifluoromethoxy)phenyl, 1-(thiazol-5-ylmethyl)-1H-pyrazol-4-yl, p-tolyl, 4-
cyclopropylphenyl, 4-(ethylsulfonyl)phenyl, 1-(6-vinylpyridin-2-yl)methyl)-1H-
pyrazol-
4-yl, 6-(benzyloxy)pyridin-3-yl, 1-(tert-butoxy-carbony1)-2,5-dihydro-1H-
pyrrol-3-yl, 1-
(2-hydroxy-1-phenylethyl)-1 H-pyrazol-4-yl, 1-(2-cyano-1-phenylethyl)-1H-
pyrazol-4-
0, 6-cyclopropylpyridin-3-yl,
4-cyano-3-methoxy-phenyl, 4-methoxy-3-
(trifluoromethyl)phenyl, 4-chlorophenyl, 1-(3,4-difluorobenzy1)-1H-pyrazol-4-
yl, 4-
methy1-3-(trifluoromethyl)phenyl, 4-
(pyrrolidine-1-carbonyl)phenyl, 4-
(isopropylarninocarbonyl)phenyl, 4-(4-rnethylpiperazin-1-yOphenyl, 3-chloro-5-
cyano-
phenyl, 3-(pyrrolidine-1-carbonyl)phenyl, 3-(methylsulfonylaminomethyl)phenyl,
3-
(1 H-pyrazol-5-yl)phenyl,
4-(methylsulfonyl)phenyl, 4-
(cyclopropylaminocarbonyl)phenyl, 1-(2-
fluoroethyl)-1H-pyrazol-4-yl, 3-
(cyclopropylmethoxy)phenyl, 3-(benzyloxy)phenyl, 3-(morpholinornethyl)phenyl,
3-
(phenoxymethyl)phenyl, 1-(3-fluoropheny1)-1H-pyrazol-4-yl, 2-cyclopropylvinyl,
6-
(trifluoromethyl)pyridin-3-yl, 1-(4-fluoropheny1)-1H-pyrazol-4-yl, 2,4-
dimethylthiazol-5-
yl,
1-propy1-1H-pyrazol-4-yl, 1-
buty1-1H-pyrazol-4-yl, 1-(2-(phenylam ino)ethyl)-1H-
pyrazol-4-yl, 4-(aminocarbonyl)phenyl, 4-(N-methylamino-carbonyl)phenyl, 3-
fluoro-4-
(N-methylaminocarbonyl)phenyl, 1-(2-(3,3-difluoroazetidin-1-ypethyl)-1H-
pyrazol-4-
1,1, 1-(2-(3,3-difluoropyrrolidin-1-ypethyl)-1H-
pyrazol-4-yl, 1424(2,2,2-
trifluoroethyl)am ino)ethyl)-1H-pyrazol-4-yl,
1-propenyl, 3-(m ethylcarbonyl-
amino)phenyl, 4-(methylsulfonylamino)phenyl, 4-(morpholine-4-carbonyl)phenyl,
4-(4-
acetylpiperazin-1-yl)phenyl, 1-(2,2-difluoroethyl)-1H-pyrazol-4-yl, 5-
isopropylfuran-2-
yl, 1-(3,3-difluorocyclopenty1)-1H-pyrazol-4-yl, 1418,3R)-3-
hydroxycyclopenty1)-1H-
pyrazol-4-yl, 1-((1S,38)-3-hydroxycyclopenty1)-1H-pyrazol-4-yl, 3-(1H-pyrazol-
4-
yl)phenyl, 5-bromofuran-2-yl, 3-(phenylamino)phenyl, 2-methylthiazol-5-yl, 3-
(phenylethynyl)phenyl, 3-phenethylphenyl, 1-(3-fluorocyclopenty1)-1H-pyrazol-4-
yl, 1-
(1-m ethoxy-2-methyl-propan-2-y1)-IH-pyrazol-4-yl,
1-(1-acryloylazetidin-3-y1)-1H-
pyrazol-4-yl, 1-(1-propionyl-azetidin-3-y1)-1H-pyrazol-4-yl, 6-oxo-1,6-
dihydropyrid in-3-
yl, 4-(piperazin-1-yl)phenyl, 1-(1-fluoro-2-methylpropan-2-y1)-1H-pyrazol-4-
yl, 3-
(trifluoromethyl)-1H-pyrazol-4-yl, 3,5-dimethylphenyl, 4-
(morpholinosulfonyl)phenyl,
3-(4-methylpiperazine-1-carbonyl)phenyl, 3-
(2-hydroxypropan-2-yl)phenyl, 1-
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
isopropy1-3-methy1-1H-pyrazol-4-yl,
1-isopropy1-5-methy1-1H-pyrazol-4-yl,
3-
cyclopropy1-1H-pyrazol-5-yl, 5-methoxycarbonylpyrrol-3-yl, 3-cyclopropy1-1-
isopropyl-
1 H-pyrazol-5-yl, 5-cyclopropy1-1-isopropy1-1H-
pyrazol-3-yl, 1-isopropy1-5-
(methoxycarbonyl)pyrrol-3-yl,
1-methy1-3-(trifluoromethyl)-1H-pyrazol-
5-yl, 1-
isopropyl-1 H-pyrazol-3-yl, 1-cyclopenty1-5-cyclopropy1-1H-pyrazol-3-yl, 1-
cyclopenty1-
3-cyclopropy1-1H-pyrazol-5-yl, 1-cyclopenty1-1H-pyrazol-3-yl, 1-isopropy1-1H-
pyrazol-
5-yl,
1-isopropyl-5-(N-methylaminocarbonyl)pyrrol-3-yl, 1-
isopropy1-5-(N,N-
dimethylamino-carbonyl)pyrrol-3-yl, 1-(2-cyclopropylethyl)-1H-pyrazol-3-yl, 1-
(2-
cyclopropylethyl)-1H-pyrazol-5-yl, 1-ethyl-1H-
pyrazol-3-yl, 3-(3, 3-dimethy1-2-
oxopyrrolidin-1-yl)phenyl, 3-(2-oxo-3-
phenylpyrrolidin-l-yl)phenyl,
styryl)phenyl, 3-(3-cyanophenyl)phenyl, 3-(3-
(methylsulfonylamino)phenyl)phenyl, 3-
(4-(methylsulfonylam ino)phenyl)phenyl,
or
methylam inosulfonyl)phenyl)phenyl.
In certain embodiments, R3 is 1 H-pyrazol-4-yl, 1-(cyclopropylmethyl)-1H-
pyrazol-4-yl,
1-(1-methylcyclopropy1)-1H-pyrazol-4-yl, 5-fluoro-1H-pyrazol-4-yl, 1-(2-
phenylpropan-
2-y1)-1H-pyrazol-4-yl, 1-(pyridin-3-y1)-1H-pyrazol-4-yl, 1-(pyridin-4-y1)-1H-
pyrazol-4-yl,
1-(pyridin-2-y1)-1H-pyrazol-4-yl, 1-[1-(N-methylam inocarbony1)-1,1-d inn
ethylm ethy1]-
1 H-pyrazol-4-yl, 5-fluoro-1-isopropy1-1H-pyrazol-4-yl,
1-(cyclopropylnriethyl)-1H-
pyrazol-5-yl, 1-(cyclopropylmethyl)-1H-pyrazol-3-yl, 1-(tetrahydro-2H-th
iopyran-4-y1)-
1 H-pyrazol-4-yl, 1-(1 , 1-d ioxidotetrahydro-2H-thiopyran-4-y1)-1H-pyrazol-4-
yl, 1-((6-(3-
oxobut-1-en-1-yl)pyridin-2-yl)methyl)-1H-pyrazol-4-yl, 3-iodophenyl, 3-methyl-
1 ,2,4-
oxadiazol-5-yl, 5-methyl-1,3,4-oxadiazol-2-yl, 1 H-im idazol-2-yl, 5-
phenyloxazol-2-yl,
1-cyclohexylpyrazol-4-yl, 1-isopropylpyrazol-4-
yl, biphenyl-3-yl,
fluorophenyl)amino)phenyl, 3-(2-oxopyrrolidin-1-yOphenyl, 3-(methylcarbonylam
ino)-
5-phenylphenyl, 3-furyl, benzofuran-3-yl, 1-pheny1-1H-pyrazol-3-yl, 5-
cyclopropylfuran-2-yl, 2-methylfuran-3-yl, 1-pheny1-1H-pyrazol-4-yl, 1-ethy1-
1H-
pyrazol-4-yl, 1-methy1-6-oxo-1,6-dihydropyridin-3-yl, furan-2-yl, 5-
phenylfuran-2-yl, 1-
isopropyl-1 H-pyrazol-4-yl, pyrimidin-5-yl, 5-methylpyridin-3-yl, 1-methy1-1H-
pyrazol-3-
yl,
4-phenylfuran-2-yl, 2-
fluorophenyl, 4-cyanophenyl, 4-m ethoxyphenyl, 4-
(trifluoromethyl)-phenyl, 4-fluorophenyl, 1-benzy1-1H-pyrazol-4-yl, 5-
chloropyridin-3-
yl, 5-fluoropyridin-3-yl, 1-methyl-1H-pyrazol-5-yl, 4-(hydroxymethyl)furan-2-
yl, 3-
cyanophenyl, 2, 5-dihydrofuran-3-yl, thiophen-3-yl, thiophen-2-yl, 1-methy1-1H-
pyrazol-4-yl, 5-methylfuran-2-yl, 5-(hydroxymethyl)furan-2-yl, 3-
(trifluoromethyl)-
71
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
phenyl, 3-methoxyphenyl, 3-fluorophenyl, pyridin-3-yl, 1-(methylsulfonyI)-1 H-
pyrazol-
4-yl, 1-cyclopenty1-1H-pyrazol-4-yl, 1-(thiophen-3-ylmethyl)-1H-pyrazol-4-yl,
4-chloro-
3-(morpholine-4-carbonyl)phenyl, 3-chloro-4-(cyclopropylaminocarbonyl)phenyl,
1-(1-
hydroxy-2-methylpropan-2-y1)-1H-pyrazol-4-yl, 1-(3-methoxybenzy1)-1H-pyrazol-4-
yl,
1-(pyridin-4-ylmethyl)-1H-pyrazol-4-yl, 1-(2-chlorobenzy1)-1H-pyrazol-4-yl, 1-
(3-
phenoxy-benzy1)-1H-pyrazol-4-yl, 1-(4-phenoxybenzy1)-1H-pyrazol-4-yl, 1-
cyclohexyl-
1 H-pyrazol-4-yl, 1-(1-phenylethyl)-1H-pyrazol-4-yl, 1-cyclobuty1-1H-pyrazol-4-
yl, 1-
(sec-buty1)-1H-pyrazol-4-yl, 4-fluoro-3-
(pyrrolidine-1-carbonyl)phenyl, 1-
(cyclopropylsulfony1)-1H-pyrazol-3-yl, 1-(cyclopropanecarbony1)-1H-pyrazol-3-
yl, 1-
(2-cyclopropylethyl)-1H-pyrazol-4-yl, 1-([1, 1'-bipheny1]-3-ylmethyl)-1H-
pyrazol-4-yl, 1-
phenethy1-1H-pyrazol-4-yl, 1-(2-
methoxybenzy1)-1H-pyrazol-4-yl, 1-(4-
rnethoxybenzy1)-1H-pyrazol-4-yl, 1-(tert-buty1)-1H-pyrazol-4-yl, 314-
dinnethylphenyl, 3-
chloro-4-ethoxyphenyl, 4-methoxy-3-methylphenyl, 2-methylbenzo[d]thiazol-5-yl,
1-
(2-phenoxybenzyI)-1 H-pyrazol-4-yl, 1-(phenylsulfony1)-1H-pyrazol-4-yl, 1-
benzoy1-1H-
pyrazol-4-yl, 1-benzhydry1-1H-pyrazol-4-yl, 1-([1,11-bipheny1]-2-ylmethyl)-1 H-
pyrazol-
4-yl, 1-(cyclohexylmethyl)-1H-pyrazol-4-yl, 1-(pyridin-3-ylmethyl)-1H-pyrazol-
4-yl,
benzofuran-2-yl, 5-ethylfuran-2-yl, 1-(2-methoxyethyl)-1H-pyrazol-4-yl, 1-
(naphthalen-
1-ylmethyl)-1H-pyrazol-4-yl, 1-([1,1'-bipheny1]-4-
ylmethyl)-1H-pyrazol-4-yl, 3-
phenoxyphenyl, 3,4-d ichlorophenyl, 3-chloro-4-methoxyphenyl, 3-methoxy-4-
methylphenyl, 1-(thiazol-4-ylmethyl)-1H-pyrazol-4-yl,
1 H-indazol-5-yl, 3,4-
dimethoxyphenyl, 4-methoxy-3,5-dimethylphenyl, 1-(oxetan-3-y1)-1H-pyrazol-4-
yl, 1-
(2-fluorobenzy1)-1H-pyrazol-4-yl, 1-(4-
fluorobenzy1)-1H-pyrazol-4-yl, 1-
(m ethoxycarbonylmethyl)-1H-pyrazol-4-yl, 1-(2-(dimethylamino)ethyl)-1H-
pyrazol-4-
yl, 3-cyano-4-methylphenyl, benzo[d][1,3]dioxo1-5-yl, 2,3-dihydrobenzofuran-5-
yl, 1-
(3-fluorobenzy1)-1H-pyrazol-4-yl, 1-(thiophen-2-ylmethyl)-1H-pyrazol-4-yl,
142,2,2-
trifluoroethyl)-1 H-pyrazol-4-yl, 1-(3-chlorobenzy1)-
1H-pyrazol-4-yl, 1-isobuty1-1H-
pyrazol-4-yl, 1-(3,3,3-trifluoropropy1)-1H-pyrazol-4-yl, 1-(d ifluoromethyl)-1
H-pyrazol-4-
YI, 1-(2-cyanoethyl)-1H-pyrazol-4-yl,
4-cyclopropylfuran-2-yl, 2,2-
difluorobenzo[d][1,3]dioxo1-5-yl, 3-fluoro-4-
(aminocarbonyl)phenyl, 3-fluoro-4-
(methylsulfonyl)phenyl, 3-chloro-4-
(trifluoromethoxy)phenyl, 5-fluoro-3-
(aminocarbonyl)phenyl, 3-(hydroxymethyl)-4-methoxyphenyl, 1-(methylsulfony1)-
1H-
pyrrol-3-yl, 1-methyl-1H-pyrrol-3-yl, 3-bromophenyl, 3-(1-methylpyrazol-4-
yl)phenyl,
3-(1-isopropylpyrazol-4-yl)phenyl, 4-phenylphenyl, 4-(4-tluoroanilino)phenyl,
3-(tert-
butoxycarbonylamino)phenyl, 1-acety1-1,2,3,6-tetrahydropyridin-4-yl, 1-
propionyl-
72
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
1 ,2,3,6-tetrahydropyridin-4-yl, 1-acryloy1-1,2,3,6-
tetrahydropyridin-4-yl, 1-methyl-
1 ,2,3,6-tetrahydropyridin-4-yl, 14(2-methylthiazol-4-yOmethyl)-1H-pyrazol-4-
yl, 1-(2-
(acetylamino)ethyl)-1H-pyrazol-4-yl,
3,5-d ich lorophenyl, 2-fluoro-4-
(methylsulfonyl)phenyl, Htert-penty1)-1H-pyrazol-4-yl, 3-(2-
morpholinoethyl)phenyl,
3-(2-(dimethylamino)ethyl)phenyl, 1-(1-(thiazol-4-yl)ethyl)-1H-pyrazol-4-yl, 1-
(tetrahydro-2H-pyran-4-y1)-1 H-pyrazol-4-yl, 3-methoxy-4-(trifluoro-
methyl)phenyl, 3-
methoxycarbony1-4-chlorophenyl, 4-
(trifluoromethoxy)phenyl, 3-methy1-4-
(trifluoromethoxy)phenyl, 4-cyclopropy1-3-(trifluoromethyl)phenyl, 2,2-
dimethy1-2, 3-
dihydrobenzofuran-5-yl, 3,5-dim ethoxyphenyl, 3,4-difluorophenyl, 4-biphenyl,
3-
chloro-5-fluorophenyl, 3,5-bis(trifluoromethyl)phenyl, 3-fluoro-5-
methoxyphenyl, 3-
(am ino-carbonyl)phenyl,
4-(cyclopropylmethoxy)phenyl, 2-fluoro-5-
(benzyloxycarbonyl)phenyl, 3-(1H-pyrazol-1-yl)phenyl, 1-(2-hydroxycyclopenty1)-
1H-
pyrazol-4-yl, 3-(N-methylam ino-sulfonyl)phenyl, 4-(2-hydroxypropan-2-
yl)phenyl, 2-
(trifluoromethyl)pyridin-4-yl, 6-phenoxypyridin-3-yl, 2-methoxypyridin-4-yl, 4-
methyl-2-
phenylthiazol-5-yl, 3-am ino-5-cyanophenyl, 1-
(tetrahydrofuran-3-yl, 3-(N-
ethylanninocarbonyl)phenyl, 3-(aminocarbonylrnethyl)phenyl, 6-phenylpyridin-3-
yl, 1-
(tetrahydro-2H-pyran-3-y1)-1H-pyrazol-4-yl, 1-(1-methoxypropan-2-y1)-1H-
pyrazol-4-
yl, 1-(2-ethoxyethyl)-1H-pyrazol-4-yl, 1-acety1-2,5-dihydro-1H-pyrrol-3-yl, 1-
acetyl-
1 ,2,5,6-tetrahydropyridin-3-yl, 1-propiony1-1,2,5,6-tetrahydropyridin-3-yl, 1-
propionyl-
2,5-d ihydro-1H-pyrrol-3-yl,
1-((1S,38)-3-hydroxycyclobuty1)-1H-pyrazol-4-
yl, 2,5-
d ihydro-1 H-pyrrol-3-yl, 1,2,5,6-
tetrahydropyridin-3-yl, 1-methy1-
1,2,5,6-
tetrahydropyridin-3-yl, 1-acryloy1-1,2,5,6-
tetrahydropyridin-3-yl, 1-acryloy1-2,5-
dihydro-1H-pyrrol-3-yl, 4-chloro-3,5-dimethylphenyl, 4-cyano-3-methylphenyl, 1-
oxo-
2,3-d ihydro-1 H-inden-5-yl,
3,4-bis(trifluoromethyl)phenyl, 3-methyl-4-
(trifluoromethyl)phenyl, 1-(benzo[b]thiophen-7-ylmethyl)-1H-pyrazol-4-yl, 4-
fluoro-3-
(N-cyclohexylaminocarbonyl)phenyl, 4-
morpholinophenyl, 4-(4-(tert-butoxy-
carbonyl)piperazin-1-yOphenyl, 3-chloro-5-methylphenyl, 3-
(methylsulfonyl)phenyl, 4-
(m ethylsulfonylam ino)-phenyl, 4-(morpholinornethyl)phenyl, 3-m
orpholinophenyl, 1-
(2-(vinylcarbonylam ino)ethyl)-1H-pyrazol-4-yl, 1-(2-am inoethyl)-1H-pyrazol-4-
yl, 3-
cyclopropy1-4-methylphenyl, 3-ethoxyphenyl, 3-(hydroxymethyl)phenyl, 1-(2-
(tert-
butoxy-carbonylamino)ethyl)-1H-pyrazol-4-yl,
3-phenethoxyphenyl, 1 ,2,3,6-
tetrahydropyrid in-4-yl, 1-(2-
(vinylsulfonylamino)ethyl)-1H-pyrazol-4-yl, 4-
(phenylamino)phenyl,
3-methyl-1H-pyrazol-4-yl, 4-(benzyloxy)phenyl, 3,5-
d ifluorophenyl,
3-fluoro-5-trifluoromethylphenyl, 3-(ethylsulfonyl)phenyl, 3-
73
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
(trifluoromethoxy)-phenyl, 1-(th iazol-5-ylmethyl)-
1H-pyrazol-4-yl, p-tolyl, 4-
cyclopropylphenyl, 4-(ethylsulfony1)-phenyl, 1-(6-vinylpyridin-2-yl)methyl)-1H-
pyrazol-
4-yl, 6-(benzyloxy)pyridin-3-yl, 1-(tert-butoxycarbony1)-2,5-dihydro-1H-pyrrol-
3-yl, 1-
(2-hydroxy-1-phenylethyl)-1 H-pyrazol-4-yl, 1-(2-cyano-1-phenylethyl)-1 H-
pyrazol-4-
yl, 6-cyclopropylpyridin-3-yl, 4-cyano-3-methoxyphenyl, 4-methoxy-3-
(trifluoromethyl)-phenyl, 4-chlorophenyl, 1-(3,4-difluorobenzy1)-1H-pyrazol-4-
yl, 4-
methy1-3-(trifluoromethyl)phenyl, 4-(pyrrolidine-1-carbonyl)phenyl, 4-
(isopropylamino-
carbony1)-phenyl, 4-(4-methylpiperazin-l-yl)phenyl, 3-chloro-5-cyanophenyl, 3-
(pyrrolid ine-1-carbonyl)phenyl, 3-(methylsulfonylam inomethyl)phenyl, 3-(1 H-
pyrazol-
5-yl)phenyl, 4-(methylsulfonyl)phenyl, 4-(cyclopropylaminocarbonyl)phenyl, 1-
(2-
fluoroethyl)-1H-pyrazol-4-yl, 3-(cyclopropylmethoxy)phenyl, 3-
(benzyloxy)phenyl, 3-
(m orpholino-m ethyl )phenyl, 3-(phenoxyrnethyl)phenyl, 1-(3-fluoropheny1)-1 H-
pyrazol-
4-yl, 2-cyclopropylvinyl, 6-(trifluoromethyl)pyridin-3-yl, 1-(4-fluoropheny1)-
1H-pyrazol-
4-yl, 2,4-dimethylthiazol-5-yl, 1-propy1-1H-pyrazol-4-yl, 1-butyl-1H-pyrazol-4-
yl, 1-(2-
(phenylamino)ethyl)-1H-pyrazol-4-yl, 4-(am inocarbonyl)phenyl, 4-(N-
methylamino-
carbonyl)phenyl, 3-fluoro-4-( N-methylarn ino-
carbonyl)phenyl, 1-(2-(3,3-
difluoroazetidin-1-ypethyl)-1H-pyrazol-4-yl, 1-(2-(3,3-difluoropyrrolidin-1-
yl)ethyl)-1H-
pyrazol-4-yl, 1-(2-(2,2,2-trifluoroethyl)amino)ethyl)-1H-pyrazol-4-yl, 1-
propenyl, 3-
(m ethylcarbonyl-am ino)phenyl, 4-(methylsulfonylamino)phenyl, 4-(morpholine-4-
carbonyl)phenyl, 4-(4-acetylpiperazin-1-yl)phenyl, 1-(2,2-difluoroethyl)-1H-
pyrazol-4-
yl, 5-isopropylfuran-2-yl, 1-(3,3-difluorocyclopenty1)-1H-pyrazol-4-yl,
141S,3R)-3-
hydroxycyclopenty1)-1H-pyrazol-4-yl, 1-((1S,3S)-3-hydroxycyclopenty1)-1 H-
pyrazol-4-
yl, 3-(1H-pyrazol-4-yl)phenyl, 5-bromofuran-2-yl, 3-(phenylamino)phenyl, 2-
methylthiazol-5-yl, 3-(phenylethynyl)phenyl,
3-phenethylphenyl, 1-(3-
fluorocyclopenty1)-1H-pyrazol-4-yl, 1-(1-methoxy-2-methyl-propan-2-y1)-1 H-
pyrazol-4-
yl, 1-(1-acryloylazetidin-3-y1)-1H-pyrazol-4-yl,
1-(1-propionyl-azetidin-3-y1)-
1H-
pyrazol-4-yl, 6-oxo-1,6-dihydropyridin-3-yl, 4-(piperazin-1-yl)phenyl, 1-(1-
fluoro-2-
methylpropan-2-y1)-1H-pyrazol-4-yl, 3-
(trifluoromethyl)-1H-pyrazol-4-yl, 3,5-
d imethylphenyl, 4-(m orphol inosulfonyl)phenyl,
3-(4-m ethylpiperazine-1-
carbonyl)phenyl, 3-(2-hydroxypropan-2-yl)phenyl, 1-isopropy1-3-methy1-1H-
pyrazol-4-
yl, 1-isopropy1-5-methy1-1H-pyrazol-4-yl,
3-cyclopropy1-1H-pyrazol-5-yl, 5-
methoxycarbonylpyrrol-3-yl, 3-cyclopropy1-1-isopropy1-1H-pyrazol-5-yl, 5-
cyclopropy1-
1-isopropy1-1H-pyrazol-3-yl, 1-isopropy1-5-(methoxycarbonyl)pyrrol-3-yl, 1-
methy1-3-
(trifluoromethyl)-1H-pyrazol-5-yl, 1-isopropy1-1H-
pyrazol-3-yl, 1-cyclopenty1-5-
74
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
cyclopropyl-1 H-pyrazol-3-yl, -1 -cyclopenty1-3-
cyclopropy1-1H-pyrazol-5-yl, 1-
cyclopentyl-1 H-pyrazol-3-yl, 1-isopropy1-1H-
pyrazol-5-yl, 1-isopropy1-5-(N-
methylaminocarbonyl)pyrrol-3-yl, 1-isopropy1-5-(N, N-dimethylamino-carbony1)-
pyrrol-
3-yl, 1-(2-cyclopropylethyl)-1H-pyrazol-3-yl, 1-(2-cyclopropylethyl)-1H-
pyrazol-5-yl, 1-
ethyl-1H-pyrazol-3-yl, 3-(3, 3-dimethy1-2-oxopynrol id in-1-
yOphenyl, 3-(2-oxo-3-
phenylpyrrolidin-1-yl)phenyl, 3-((E)-styryl)phenyl, 3-(3-cyanophenyl)phenyl, 3-
(3-
(methylsulfonylamino)phenyl)phenyl, 3-(4-(methylsulfonylamino)phenyl)phenyl,
or 3-
(4-(N-methylaminosulfonyl)phenyl)phenyl.
In certain embodiments, R3 is aryl or heteroaryl, wherein each aryl and
heteroaryl is
optionally substituted with one or more groups Rx; provided R3 is not phenyl,
fluorophenyl, chlorophenyl, pyridyl, nitrophenyl, or propylisoxazole.
In certain embodiments, R3 is pyrazol-4-yl, optionally substituted with Rx.
In certain embodiments, Rx is C1-6 alkyl, that is substituted with one or more
groups
independently selected from Rxa, oxo, halo, -NO2, -N(Rv)2, -CN, -C(0)-
N(Rv)2, -s(o)-N(Rv)2, -S(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-C(0)-Rv, -
C(0)-Rv, -C(0)-0-Rv, -S(0)-Rv, -S(0)2-Rv, -C(0)-N(Rv)2, -S(0)2-
N(Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-C(0)-0Rv, -N(Rv)-S(0)-Rv, and -
N(Rv)-8(0)2-Rv.
In certain embodiments, Rx is C1-6alkyl that is optionally substituted with
Rxa.
In certain embodiments, R3 is pyrazol-4-yl, substituted with Rx.
In certain embodiments, R3 is phenyl that is substituted with oxo, C1-6 alkyl,
C2-6
alkenyl, C2-6a1kyny1, carbocyclyl, aryl, heteroaryl, heterocycle, -N(Rv)2, -
CN, -
C(0)-N(Rv)2, -S(0)-N(Rv)2, -S(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-
C(0)-Rv, -0-C(0)-0-Rv, -C(0)-Rv, -C(0)-0-Rv, -S(0)-Rv, -
S(0)2-Rv, -0-C(0)-N(Rv)2, -N(Rv)-C(0)-0Rv, -N(Rv)-C(0)-N(Rv)2,
-S(0 )2-N( Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-S(0)-Rv, -N(Rv)-S(0)2-Rv,
-N(Rv)-S(0)-N(Rv)2, or -N(Rv)-S(0)2-N(Rv)2, wherein any C1-6 alkyl, C2-6
alkenyl, C2-6 alkynyl, carbocyclyl, aryl, heteroaryl, and heterocycle is
optionally
substituted with one or more groups independently selected from Rxa, oxo,
halo, -
NO2, -N(Rv)2, -CN, -C(0)-N(Rv)2, -S(0)-N(Rv)2, -8(0)2-N(Rv)2, -0-
Rv, -S-Rv, -0-C(0)-Rv, -C(0)-Rv, -C(0)-0-Rv, -8(0)-Rv, -
S(0)2-Rv, -C(0)-N(Rv)2, -S(0)2-N(Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
C(0)-0Rv, -N(Rv)-S(0)-Rv, -N(Rv)-S(0)2-Rv, or C1-6alkyl that is optionally
substituted with one or more groups independently selected from oxo and halo.
In certain embodiments, Rx is C2-6 alkenyl or C2-6 alkynyl, wherein any C2-6
alkenyl
and C2-6a1kyny1 is optionally substituted with one or more groups
independently
selected from Rxa, oxo, halo, -NO2, -N(Rv)2, -CN, -C(0)-N(Rv)2, -8(0)-
N(Rv)2, -S(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-C(0)-Rv, -C(0)-Rv, -
C(0)-0-Rv, -8(0)-Rv, -S(0)2-Rv, -C(0)-N(Rv)2, -3(0)2-N(Rv)2, -
N(Rv)-C(0)-Rv, -N(Rv)-C(0)-0Rv, -N(Rv)-8(0)-Rv, and -N(Rv)-
S(0)2-Rv
In certain embodiments, Rx is selected from C2-6 alkenyl, C2-6 alkynyl,
carbocyclyl,
aryl, heteroaryl, heterocycle, -F, -CI, -Br, -I, -NO2, -N(Rv)2, -CN, -C(0)-
N(Rv)2, -S(0)-N(Rv)2, -8(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-C(0)-Rv, -
0-C(0)-0-Rv, -C(0)-Rv, -C(0)-0-Rv, -S(0)-Rv, -S(0)2-Rv, -0-
C(0)-N(Rv)2, -N(Rv)-C(0)-0Rv, -N(Rv)-C(0)-N(Rv)2, -S(0)2-N(Rv)2,
-N(Rv)-C(0)-Rv, -N(Rv)-S(0)-Rv, -N(Rv)-S(0)2-Rv, -N(Rv)-8(0)-
N(Rv)2, and -N(Rv)-S(0)2-N(Rv)2, wherein any C2-6 alkenyl, C2-6 alkynyl,
carbocyclyl, aryl, heteroaryl, and heterocycle is optionally substituted with
one or more
groups independently selected from Rxa, oxo, halo, -NO2, -N(Rv)2, -CN, -
C(0)-N(Rv)2, -8(0)-N(Rv)2, -S(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-
C(0)-Rv, -C(0)-Rv, -C(0)-0-Rv, -8(0)-Rv, -S(0)2-Rv, -C(0)-
N(Rv)2, -S(0)2-N(Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-C(0)-0Rv, -N(Rv)-
S(0)-Rv, -N(Rv)-S(0)2-Rv, and C1-6 alkyl that is optionally substituted with
one
or more groups independently selected from oxo and halo.
In certain embodiments, R3 is heteroaryl that is substituted with oxo, C1-6
alkyl, C2-6
alkenyl, C2-6 alkynyl, carbocyclyl, aryl, heteroaryl, heterocycle, -F, -Cl, -
Br, -I,
-NO2, -N(Rv)2, -CN, -C(0)-N(Rv)2, -8(0)-14(Rv)2, -8(0)2-N(Rv)2, -
0-Rv, -S-Rv, -0-C(0)-Rv, -0-C(0)-0-Rv, -C(0)-Rv, -C(0)-0-
Rv, -8(0)-Rv, -S(0)2-Rv, -0-C(0)-N(Rv)2, -N(Rv)-C(0)-0Rv, -
N(Rv)-C(0)-N(Rv)2, -S(0)2-N(Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-S(0)-Rv,
-N(Rv)-S(0)2-Rv, -N(Rv)-S(0)-N(Rv)2, or -N(Rv)-S(0)2-N(Rv)2;
wherein any C1-6 alkyl is substituted with one or more groups independently
selected
from Rxa, oxo, halo, -NO2, -N(Rv)2, -CN, -C(0)-N(Rv)2, -S(0)-N(Rv)2, -
S(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-C(0)-Rv, -C(0)-Rv, -C(0)-0-Rv,
76
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
-S(0)-Rv, -8(0)2-Rv, -C(0)-N(Rv)2, -S(0)2-N(Rv)2, -N(Rv)-C(0)-
Rv, -N(Rv)-C(0)-0Rv, -N(Rv)-S(0)-Rv, and -N(Rv)-8(0)2-Rv; and
wherein any C2-6alkenyl, C2-6alkynyl, carbocyclyl, aryl, heteroaryl, and
heterocycle
is optionally substituted with one or more groups independently selected from
Rxa,
oxo, halo, -NO2, -N(Rv)2, -CN, -C(0)-N(Rv)2, -8(0)-N(Rv)2, -8(0)2-
N(Rv)2, -0-Rv, -S-Rv, -0-C(0)-Rv, -C(0)-Rv, -C(0)-0-Rv, -
S(0)-Rv, -8(0)2-Rv, -C(0)-N(Rv)2, -S(0)2-N(Rv)2, -N(Rv)-C(0)-Rv,
-N(Rv)-C(0)-0Rv, -N(Rv)-S(0)-Rv, -N(Rv)-S(0)2-Rv, and C1-6 alkyl
that is optionally substituted with one or more groups independently selected
from oxo
and halo.
In certain embodiments, R3 is a 5-membered heteroaryl that is substituted with
oxo,
C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, carbocyclyl, aryl, heteroaryl,
heterocycle, -F,
-Cl, -Br, -I, -NO2, -N(Rv)2, -CN, -C(0)-N(Rv)2, -S(0)-N(Rv)2, -
S(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-C(0)-Rv, -0-C(0)-0-Rv, -C(0)-
Rv, -C(0)-0-Rv, -8(0)-Rv, -3(0)2-Rv, -0-C(0)-N(Rv)2, -N(Rv)-
C(0)-0Rv, -N(Rv)-C(0)-N(Rv)2, -8(0)2-N(Rv)2, -N(Rv)-C(0)-Rv, -
N(Rv)-8(0)-Rv, -N(Rv)-8(0)2-Rv, -N(Rv)-S(0)-N(Rv)2, or -N(Rv)-
S(0)2-N(Rv)2; wherein any C1-6alkyl, is substituted with one or more groups
independently selected from Rxa, oxo, halo, -NO2, -N(Rv)2, -CN, -C(0)-
N(Rv)2, -S(0)-N(Rv)2, -S(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-C(0)-Rv, -
C(0)-Rv, -C(0)-0-Rv, -S(0)-Rv, -8(0)2-Rv, -C(0)-N(Rv)2, -8(0)2-
N(Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-C(0)-0Rv, -N(Rv)-S(0)-Rv, and -
N(Rv)-S(0)2-Rv; and wherein any C2-6alkenyl, C2-6alkynyl, carbocyclyl, aryl,
heteroaryl, and heterocycle is optionally substituted with one or more groups
independently selected from Rxa, oxo, halo, -NO2, -N(Rv)2, -CN, -C(0)-
N(Rv)2, -8(0)-N(Rv)2, -8(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-C(0)-Rv, -
C(0)-Rv, -C(0)-0-Rv, -S(0)-Rv, -S(0)2-Rv, -C(0)-N(Rv)2, -8(0)2-
N(Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-C(0)-0Rv, -N(Rv)-S(0)-Rv, -N(Rv)-
S(0)2-Rv, and C1-6 alkyl that is optionally substituted with one or more
groups
independently selected from oxo and halo.
In certain embodiments, R3 is phenyl that is substituted with oxo, C1-6 alkyl,
C2-6
alkenyl, C2-6 alkynyl, carbocyclyl, aryl, heteroaryl, heterocycle, -N(Rv)2, -
CN, -
C(0)-N(Rv)2, -S(0)-N(Rv)2, -8(0)2-N(Rv)2, -0-Rv, -S-Rv, -0-
77
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
C(0)-RV, -0-C( 0)-0-Rv, -C(0 )-Rv, -C(0)-0-Rv, -5( 0)-Rv, -
S(0)2-Rv, -0-C(0)-N(Rv)2, -N(Rv)-C(0)-0Rv, -N(Rv)-C(0)-N(Rv)2,
-S(0 )2-N( Rv)2, -N( Rv)-C(0)-Rv, -N(Rv)-S(0)-Rv, -N(Rv)-S(0)2-Rv,
-N(Rv)-S(0)-N(Rv)2, or -N(Rv)-S(0)2-N(Rv)2; wherein any C1-6 alkyl, is
substituted with one or more groups independently selected from Rxa, oxo,
halo, -
NO2, -N(Rv)2, -CN, -C(0)-N(Rv)2, -S(0)-N(Rv)2, -S(0)2-N(Rv)2, -0-
Rv, -S-Rv, -0-C(0)-Rv, -C(0)-Rv, -C(0)-0-Rv, -S(0)-Rv, -
S(0)2-Rv, -C(0)-N(Rv)2, -S(0)2-N(Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-
C(0)-0Rv, -N(Rv)-8(0)-Rv, and -N(Rv)-8(0)2-Rv; and wherein any C2-6
alkenyl, C2-6 alkynyl, carbocyclyl, aryl, heteroaryl, and heterocycle is
optionally
substituted with one or more groups independently selected from Rxa, oxo,
halo, -
NO2, -N(Rv)2, -CN, -C(0)-N(Rv)2, -8(0)-N(Rv)2, -8(0)2-N(Rv)2, -0-
Rv, -S-Rv, -0-C(0)-Rv, -C(0)-Rv, -C(0)-0-Rv, -S(0)-Rv, -
S(0)2-Rv, -C(0)-N(Rv)2, -S(0)2-N(Rv)2, -N(Rv)-C(0)-Rv, -N(Rv)-
C(0)-0Rv, -N(Rv)-S(0)-Rv, -N(Rv)-S(0)2-Rv, and C1-6alkyl that is
optionally substituted with one or more groups independently selected from oxo
and
halo.
In certain embodiments, R2 and R3 taken together with the atoms to which they
are
attached form a cyclohexyl ring, which is optionally substituted with one or
more
groups Rx.
In certain embodiments, R2 and R3 taken together with the atoms to which they
are
attached form a phenyl ring, which is optionally substituted with one or more
groups
Rx.
In certain embodiments, R4 is H, methyl, ethyl, propyl, cyclopropylmethyl, 2-
hydroxyethyl, 2-(dinnethylannino)ethyl, phenyl, benzyl, or 2-nnethoxyethyl.
In certain embodiments, R4 and R3 taken together with the atoms to which they
are
attached form a heterocyclyl.
In certain embodiments, the compound is other than any one of the following:
78
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
I0
N(2
0
11
0
0
yao,
TT
KC 1=C
)-E1
NC
0
=
NC
0
11
NC
0
0
) 91,Th I 0
NC
0
or
y_11,N
11
NC
0
0
I
NC
79
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In other embodiments, the agent is a (small molecule) inhibitor of jumonji at-
rich
interactive domain la (jaridl a) and lb (jaridl b) histone demethylase, such
as
described in PCT publication W02014055634A1 (incorporated by reference
herein).
In another embodiment, the JIVIJD3 demethylase inhibitor comprises any of the
N-2-
(2-pyridinyI)-4-pyrimidinyl-beta-alanine derivatives as disclosed in
W02012052390 to
Atkinson et at, which is hereby incorporated by reference in its entirety.
Exemplary
inhibitors include those having the structure of:
Ri ,.......... ..
NH (CH 2)2C0 OH
I
"........ci
I
R2
R3
where R1 is C1-6 alkyl; C3-7 cycloalkyl; C1-6 haloalkyl; a 5, 6 or 7-membered
aryl or
heteroaryl (which heteroaryl contains one or more heteroatoms selected from N,
0
and S and which is optionally fused to phenyl), said 5, 6 or 7-membered aryl
or
heteroaryl being optionally substituted with one or more substituents
independently
selected from C1-3alkyl; 0¨C1-6a1ky1 (which is optionally substituted by
phenyl or
naphthyl, each of which may be substituted by one of more substituents
independently
selected from halo); ¨0-cyclohexyl (which is optionally fused with phenyl);
C(0)NRc
2; or NRaRb, each Ra and Rb is independently selected from: H; C1-3alkyl which
is
optionally substituted by one or more substituents independently selected from
phenyl
(which phenyl is optionally substituted by one or more substituents
independently
selected from C1-3a1ky1, 0¨C1-3a1ky1, C(0)NRc 2, halo and cyano), C(0)NRc 2,
a4,
5, 6 or 7-membered heterocyclic or heteroaryl group (containing one or more
heteroatoms independently selected from N, 0 and, S), a 3, 4, 5, 6 or 7-
membered
cycloalkyl group (which is optionally fused to phenyl), halo, 0C1-3alkyl, OH,
¨
NHCOC1-3a1ky1 NRc 2 and C(0)NHCH2C(0)NRc 2; a 3, 4, 5, 6 or 7-membered
cycloalkyl group (which is optionally fused to phenyl), or Ra and Rb together
form a 5,
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
6 or 7-membered heterocyclic group optionally containing one or more further
heteroatoms independently selected from N, 0, S or 5(0)2 said heterocyclic
group
being optionally fused to a 5, 6 or 7-membered aryl or heteroaryl ring
containing one
or more heteroatoms independently selected from N, 0 and S; the heterocylic
ring
and/or the aryl or heteroaryl to which it is optionally fused being optionally
substituted
by one or more substituents independently selected from halo, OH, C1-3a1ky1,
0¨C1-
3a1ky1, C(0)C1-3alkyl, S(0)2C1-3alkyl, NHC(0)C1-3a1ky1, NHS(0)2C1-3a1ky1,
C(0)NRc 2, C(0)NRd 2 (wherein Rd and Rd together form a 5 or 6-membered
heterocylic ring), NRC 2C(0)phenyl, S(0)2NRc 2, =0(oxo) and 5, 6 or 7-membered
aryl or heteroaryl (containing one or more heteroatoms independently selected
from
N, 0 and S); R2 and R3 are each independently selected from: H, (CH2)1-
3NRc(CH2)1-3NRc 2, (CH2)1-6NRc 2; C1-3 alkyl; 0¨C1-3a1ky1; C1-3ha1oa1ky1;
(CH2)0-3NRaRb (wherein Ra and Rb are as defined above); (CH2)0-3NHPh; (CH2)0-
30Ph; (CH2)0-3Ph; or R2 and R3 together form a fused phenyl ring, and each Rc
is
independently selected from hydrogen and C1-3a1ky1 or a pharmaceutically
acceptable salt thereof. Suitable inhibitors of this structure include,
without limitation,
N-[6-( 1,1-dim ethylethyl)-2-(2-pyridiny1)-4-pyrim id iny1]-13-alan ine; N-[2-
(2-pyridiny1)-6-
(trifluoromethyl)-4-pyrim idiny1]-13-alanine;
N-[6-(4-morpholiny1)-2-(2-
pyridiny1)-4-
pyrim idinyli-p-alanine; N-[6-(methylam ino)-2-(2-pyridiny1)-4-pyrim idiny1]-
I3-alanine; N-
[2-(2-pyridiny1)-6-(1-pyrrolidiny1)-4-pyrimidinyl]-13-alanine;
N-[6-[(2-
hydroxyethyl)am ino]-2-(2-pyridiny1)-4-pyrimidiny11-13-alanine;
N-[6-
[(phenyl methyl)amino]-2-(2-pyrid iny1)-4-pyrim idiny1]-13-alanine;
N-[6-[(2-
hydroxyethyl)(methyl)am ino]-2-(2-pyridiny1)-4-pyrim idiny1]-13-alanine;
N-[6-
(dimethylam ino)-2-(2-pyridinyI)-4-pyrim id iny1]-13-alan ine;
and N-[2-(2-pyridiny1)-6-
(1 ,2,4,5-tetrahydro-3/-/-3-benzazepin-3-y1)-4-pyrim id iny1]-13-alanine.
Other JMJ D3
demethylase inhibitors disclosed in W02012052390 to Atkinson et at, which is
hereby
incorporated by reference in its entirety, are also suitable for use in the
methods as
described herein.
In another embodiment, the JMJD3 demethylase inhibitor comprises any one of
the
small molecule JMJD3 inhibitors disclosed in W02013143597 to Barker et at,
which
is hereby incorporated by reference in its entirety. Exemplary inhibitors
include those
having the structure of
81
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
COOH
...e.õ,..X
1
N
where X is ¨(R1)o-i-(R2)o-i-R3 or¨RI¨R4; Each R1 is independently NH, N(01-
13),
0; R2 is a linker group with a maximum length of 5 atoms between R1 and R3 and
is
selected from: ¨CO¨C1-6 alkyl-, ¨CO¨, ¨CO¨C1-6 alkyl-O¨, ¨CO¨C1-6
alkyl-S¨, ¨CO¨C1-6 alkyl-0¨ C1-6 alkyl-, ¨C1-6 alkyl-, C1-6a1ky1-0¨, ¨C1-
6a1ky1-8 02¨, ¨C 1-6alkyl-N H¨C 0¨, ¨C1-3 alkyl-C3-6 cycloalkyl-C 1-3alky1-0¨
wherein each alkyl is straight chain or branched and may be optionally
substituted by
one or more substituents independently selected from phenyl or ¨OH; R3 is
selected
from: a C6-12 mono or bicyclic aryl group, (each of which may be optionally
substituted
one or more times by substituents independently selected from halo, C1-6alkyl,
C1-6
haloalkyl, C1-6a1koxy, NHCOC1-3a1ky1, ¨0¨ phenyl, ¨CH2-phenyl, phenyl
(optionally substituted by C1-3alkyl), OH, NH2, CONH2, CN, ¨NHCOC1 -3alkyINH2,
¨HCOC1-3a1ky1, NHCOOC 1-3alkyl, ¨NHSO2C1-3alkyl, ¨802C 1-3alkyl or
¨NHCOC1-3alkyl-NHCOC1-4 alkyl
0
7µN.
NH
)%1IN 7C
S
)-
a 5-12 membered mono or bicyclic heteroaryl group (optionally substituted by
one or
more substituents independently selected from phenyl, CH2phenyl, ¨C1-6 alkyl, -
oxo), a 5 or 6 membered heterocyclic group containing one or more
heteromoieties
independently selected from N, S, SO, SO2 or 0 and optionally fused to a
phenyl
group (optionally substituted by one or more substituents independently
selected from
phenyl, CH2phenyl, C1-6 alkyl), or a 3-7 membered cycloalkyl (including
bridged
82
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
cycloalkyl) and optionally fused to a phenyl group (and optionally substituted
by one
or more substituents independently selected from OH, phenyl, CH2 phenyl), R4
is
selected from: C1-6 straight chain or branched alkyl (optionally substituted
by NH2),
COC1-6 straight chain or branched alkyl. Suitable inhibitors of this structure
include,
without limitation, 3-{[(4-chlorophenyl)acetyliamino}-4-pyridinecarboxylic
acid; 3-{[(4-
methylphenyl)acetyl]am ino}-4-pyridinecarboxylic acid; -[(3-
phenylpropanoyl)amino]-4-
pyridinecarboxylic acid; -[(phenylcarbonyl)amino]-4-pyridinecarboxylic acid; -
[(2,2-
dimethylpropanoyDamino]-4-pyridinecarboxylic acid; -{[(phenyloxy)acetyl]amino)-
4-
pyridinecarboxylic acid; -{[4-(4-methylphenyl)butanoyl]amino}-4-
pyridinecarboxylic
acid; -[(2-naphthalenylacetyl)amino]-4-pyridinecarboxylic acid;
naphthalenyObutanoynamino}-4-pyridinecarboxylic
acid; and 4[444-
bronnophenyl)butanoyl]ann ino}-4-pyridinecarboxylic acid. Additional small
molecule
JMJD3 inhibitors disclosed in W02013143597 to Barker et at, which is hereby
incorporated by reference in its entirety, are also suitable for use in the
methods
described herein.
Small molecule JMJD3 demethylase inhibitors can be readily modified using
techniques known in the art to increase bioavailability (see Hetal et al, "A
Review on
Techniques for Oral Bioavailability Enhancement of Drugs," Intl. J. Pharm.
Sci. Rev.
Res. 4(3): 203-223 (2010) and Huttunen et at, "Prodrugs¨from Serendipity to
Rational Design," Pharmacol. Rev. 63(3):750-771 (2011), which are hereby
incorporated by reference in their entirety). For example, common
modifications to
increase the solubility and dissolution rate of small molecules include
particle size
reduction, modification of the crystal habit, dispersion in carriers,
inclusion
complexation, salt formation, and change in pH. Modification of the small
molecule
into a prodrug form using, for example, attached ionizable or polar neutral
groups (e.g.,
phosphate esters, amino acids, sugar moieties) is also known to enhance
solubility
and dissolution rate. Common modification to increase permeability and
absorption
include, for example, conversion of hydrophilic hydroxyl, thiol, carboxyl,
phosphate, or
amine groups to more lipophilic alkyl or aryl esters.
In another embodiment, the JMJD3 demethylase inhibitor is a JMJD3 antisense
RNA,
shRNA, or siRNA oligonucleotide.
The use of antisense methods to inhibit the in vivo translation of genes and
subsequent
protein expression is well known in the art (e.g., U.S. Pat. No. 7,425,544 to
Dobie et
83
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
at; U.S. Pat. No. 7,307,069 to Karras et at; U.S. Pat. No. 7,288,530 to
Bennett et at;
U.S. Pat. No. 7,179,796 to Cowsert et at, which are hereby incorporated by
reference
in their entirety). Antisense nucleic acids are nucleic acid molecules (e.g.,
molecules
containing DNA nucleotides, RNA nucleotides, or modifications (e.g.,
modification that
increase the stability of the molecule, such as 2'-0-alkyl (e.g., methyl)
substituted
nucleotides) or combinations thereof) that are complementary to, or that
hybridize to,
at least a portion of a specific nucleic acid molecule, such as an mRNA
molecule (see
e.g., Weintraub, H. M., "Antisense DNA and RNA," Scientific Am. 262:40-46
(1990),
which is hereby incorporated by reference in its entirety). The antisense
nucleic acid
molecule hybridizes to its corresponding target nucleic acid molecule, such as
JMJ D3,
to form a double-stranded molecule, which interferes with translation of the m
RNA, as
the cell will not translate a double-stranded mRNA. Antisense nucleic acids
used in
the methods of the present disclosure are typically at least 10-12 nucleotides
in length,
for example, at least 15, 20, 25, 50, 75, or 100 nucleotides in length. The
antisense
nucleic acid can also be as long as the target nucleic acid with which it is
intended to
form an inhibitory duplex. Antisense nucleic acids can be introduced into
cells as
antisense oligonucleotides, or can be produced in a cell in which a nucleic
acid
encoding the antisense nucleic acid has been introduced, for example, using
gene
therapy methods.
siRNAs are double stranded synthetic RNA molecules approximately 20-25
nucleotides in length with short 2-3 nucleotide 3' overhangs on both ends. The
double
stranded siRNA molecule represents the sense and anti-sense strand of a
portion of
the target mRNA molecule, in this case a portion of the JMJD3 nucleotide
sequence
(SEQ ID NO: 1). siRNA molecules are typically designed to target a region of
the
mRNA target approximately 50-100 nucleotides downstream from the start codon.
Upon introduction into a cell, the siRNA complex triggers the endogenous RNA
interference (RNAi) pathway, resulting in the cleavage and degradation of the
target
mRNA molecule. Various improvements of siRNA compositions, such as the
incorporation of modified nucleosides or motifs into one or both strands of
the siRNA
molecule to enhance stability, specificity, and efficacy, have been described
and are
suitable for use in accordance with this aspect of the disclosure (see e.g.,
W02004/015107 to Giese et at; W02003/070918 to McSwiggen et at;
84
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
W01998/39352 to Imanishi et at; U.S. Patent Application Publication No.
2002/0068708 to Jesper etal.; U.S. Patent Application Publication No.
2002/0147332
to Kaneko et al; U.S. Patent Application Publication No. 2008/0119427 to Bhat
et at,
which are hereby incorporated by reference in their entirety).
Short or small hairpin RNA molecules are similar to siRNA molecules in
function, but
comprise longer RNA sequences that make a tight hairpin turn. shRNA is cleaved
by
cellular machinery into siRNA and gene expression is silenced via the cellular
RNA
interference pathway. shRNA molecules that effectively interfere with JMJD3
expression have been developed, as described herein, and have the following
nucleic
acid sequences:
51-
CAGGGAAGTTTCGAGAAGTCCTATAGTGAAGCCACAGATGTATAGGACTCTCGA
AC TTCCCTT-3' (SEQ ID NO: 1) and
5'-
ACACCAGCAGTAGCAACAGCAATAGTGAAGCCACAGATGTATTGCTGTTGCTAC
TG CTGGTGG-3' (SEQ ID NO: 2)
b. EGFR/HER2 1nhibitorsHistone Demethylase Inhibitors
In certain embodiments, the anti-PESC agent is a receptor tyrosine kinase
inhibitor,
and is preferably an EGFR inhibitor, a HER2 inhibitor or a dual EGFR/HER2
inhibitor.
Exemplary EGFR inhibitors/antagonists include, inter alia, small-molecule EGFR
inhibitors/antagonists, such as gefitinib, erlotinib, lapatinib, afatinib
(also referred to as
BIBVV2992), neratinib, ABT-414, dacomitinib (also referred to as PF-00299804),
AV-
412, PD 153035, vandetanib, PKI-166, pelitinib (also referred to as EKB-569),
canertinib (also referred to as CI-1033), icotinib, poziotinib (also referred
to as
NOV120101), BMS-690514, CUDC-101, AP26113 or XL647.
In certain embodiments, the anti-PESC agent is an EGFR tyrosine kinase
inhibitor
(EGFR-TKI). Exemplary EGFR-TKI include afatinib, erlotinib, gefitinib,
neratinib,
dacomitinib and osimertinib.
VVZ8040 is a novel mutant-selective irreversible EGFRT790M inhibitor, does not
inhibit
ERBB2 phosphorylation (T798I).
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Protea some Inhibitors
In other embodiments, the anti-PESC agent can be a proteasome inhibitor.
The proteasome inhibitor may be any proteasome inhibitor known in the art. In
particular, it is one of the proteasome inhibitors described in more detail in
the following
paragraphs.
Suitable proteasome inhibitors for use in combinations described herein
include (a)
peptide boronates, such as bortezomib (also known as Velcaden" and PS341),
delanzomib (also known as CEP-18770), ixazomib(also known as MLN9708)
or ixazomib citrate; (b) peptide aldehydes, such as MG132 (Z-Leu-Leu-Leu-H),
MG115 (Z-Leu-Leu-Nva-H), IPSI 001, fellutamide B, ALLN (Ac-Leu-Leu-N1e-H, also
referred to as calpain inhibitor I), and leupeptin (Ac-Leu-Leu-Arg-al); (c)
peptide vinyl
sulfones, (d) epoxyketones, such as epoxomicin, oprozomib (also referred to as
PR-
047 or ONX 0912), PR-957 (also known as ONX 0914), and carfilzomib (also
referred
to as PR-171); and (e) 13-lactones, such as lactacystin, omuralide,
salinosporamide A
(also known as NPI-0052 and marizomib), salinosporamide B, belactosines,
cinnabaramides, polyphenols, TMC-95, and P5-519.
In a preferred embodiment, the proteasome inhibitor is bortezomib, also known
as
VELCADE and P8341. In a preferred embodiment, the proteasome inhibitor is
[(1R)-
3-methyl-1-[[(25)-3-phenyl-2-(pyrazine-2-
carbonylamino)propanoyl]amino]butyl]boronic acid, or a pharmaceutically
acceptable
salt, solvate, hydrate, cocrystal, or prodrug thereof. In a preferred
embodiment, the
proteasome inhibitor is the compound of Formula (44):
Formula (44)
0
,IrC I
HO
I H
OH 0
or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug
thereof. Bortezonnib is commercially available.
86
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In a preferred embodiment, the proteasome inhibitor is carfilzomib, also known
as PR-
171 or KYPROLIS. In a preferred embodiment, the proteasome inhibitor is (2S )-
4-
methyl-N-[(25)-1-[[(2S)-4-methyl-1-[(2 R)-2-m ethyloxiran-2-yI]-1-oxopentan-2-
yliam ino]-1-oxo-3-phenylpropan-2-yI]-2-[[(2S)-2-[(2-morpholin-4-ylacetyl)am
ino]-4-
phenylbutanoyljaminoThentanamide, or a pharmaceutically acceptable salt,
solvate,
hydrate, cocrystal, or prodrug thereof In a preferred embodiment, the
proteasome
inhibitor is the compound of Formula (45):
Formula (45)
0
0
H
N
CwTh
0
0
or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug
10 thereof. Carfilzomib is commercially available.
In a preferred embodiment, the proteasome inhibitor is delanzomib, also known
as
CEP-18770. In a preferred embodiment, the proteasome inhibitor is [(1R)-1 -
[[(2S,3R)-
3-hydroxy-2-[(6-phenylpyridine-2-carbonyl)am ino]butanoyliam ino]-3-
methylbutyl]boronic acid, or a pharmaceutically acceptable salt, solvate,
hydrate,
15 cocrystal, or prodrug thereof. In a preferred embodiment, the
proteasome inhibitor is
the compound of Formula (46):
Formula (46)
HO
OH
0
B
0
"rti
OH
or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug
thereof.
In a preferred embodiment, the proteasome inhibitor is ixazomib, also known as
MLN-
20 9708 or ixazomib citrate. In a preferred embodiment, the
proteasome inhibitor is 4-
(carboxym ethyl)-24( R)-1-(2-(2,5-dichlorobenzam ido)acetam ido)-3-
methylbutyI)-6-
oxo-1, 3,2-dioxaborinane-4-carboxylic acid, or a pharmaceutically acceptable
salt,
solvate, hydrate, cocrystal, or prodrug thereof. In a preferred embodiment,
the
87
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
proteasome inhibitor is 1,3,2-dioxaborolane-4,4-diacetic acid, 2-[(1R)-1-[[2-
[(2,5-
dichlorobenzoyDamino] acetyl] amino]-3-methylbutyI]-5-oxo-, or a
pharmaceutically
acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof. In a
preferred
embodiment, the proteasome
inhibitor is 2,7-(2-[(1R)-1 -{[N-
(2,5-
dichlorobenzoyl)glycyl]am ino)-3-methylbuty1]-5-oxo-1,3,2-dioxaborolane-4,4-
diylidiacetic acid, or a pharmaceutically acceptable salt, solvate, hydrate,
cocrystal, or
prodrug thereof. In a preferred embodiment, the proteasome inhibitor is the
compound
of Formula (47):
Formula (47)
0
0
0
HO
CI 0
0
0
7
CI
0
or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug
thereof.
In a preferred embodiment, the proteasome inhibitor is 1B4(1R)-142-(2,5-
dichlorobenzamido)acetarnido]-3-nnethylbutyl}boronic acid, or a
pharmaceutically
acceptable salt, solvate, hydrate, cocrystal, or prodrug thereof In a
preferred
embodiment, the proteasome inhibitor is the compound of Formula (48):
Formula (48)
CI HO
OH
0
B
-
N
CI
0
or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug
thereof. Ixazomib citrate is commercially available.
In a preferred embodiment, the proteasome inhibitor is marizomib, also known
as NP I-
0052 and Salinosporamide A. In a preferred embodiment, the proteasome
inhibitor is
(4R, 58 )-4-(2-chloroethyl)-1-((1S)-cyclohex-2-enyl(hydroxy)m ethyl)-5-methy1-
6-oxa-2-
azabicyclo[3.2.0]heptane-3,7-dione, or a pharmaceutically acceptable salt,
solvate,
hydrate, cocrystal, or prodrug thereof In a preferred embodiment, the
proteasome
inhibitor is the compound of Formula (49):
88
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Formula (49)
.....
OH
0,
Cl
or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug
thereof.
In a preferred embodiment, the proteasome inhibitor is oprozimib, also known
as PR-
047 or ONX 0912. In a preferred embodiment, the proteasome inhibitor is N-
[(2S)-3-
methoxy-1-[[(2S)-3-methoxy-1-[[(2S)-1-[(2R)-2-methyloxiran-2-y1]-1-oxo-3-
phenylpropan-2-yl]am ino]-1-oxopropan-2-yl]am ino]-1-oxopropan-2-yI]-2-methyl-
1 ,
thiazole-5-carboxam ide, or a pharmaceutically acceptable salt, solvate,
hydrate,
cocrystal, or prodrug thereof. In a preferred embodiment, the proteasome
inhibitor is
the compound of Formula (50):
Formula (50)
=0
0 0
N
H 0
0
or a pharmaceutically acceptable salt, solvate, hydrate, cocrystal, or prodrug
thereof.
Exemplary proteasome inhibitors include bortezomib, carfilzomib, ixazomib,
oprozomib or marizomib
CEP-18770, disulfiram, epigallocatechin-3-gallate, epoxomicin, lactacystin,
MG132,
MLN9708, ONX 0912, and salinosporannide A.
In other embodiments, the anti-P ESC agent is Delanzomib (CEP-18770), an
orally
active inhibitor of the chymotrypsin-like activity of proteasome with IC50 of
3.8 nM, with
only marginal inhibition of the tryptic and peptidylglutannyl activities of
the proteosonne.
89
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
In other embodiments, the anti-PESC agent is CEP-18770, an novel orally-active
inhibitor of the chymotrypsin-like activity of the proteasome with a cellular
IC50 value
of 3.8 nM.
d. STAT3 Inhibitors
In certain embodiments, the anti-PESC agent is a STAT3 inhibitor such as
Stattic.
Stattic is nonpeptidic small molecule that potently inhibits STAT3 activation
and
nuclear translocation with 1C5o of 5.1 pM in cell-free assays, highly
selectivity over
STAT1.
Non-limiting examples of STAT3 inhibitors include BP-1-102, S3I-M2001, STA-21,
531- 201, Galiellalactone, a polypeptide having the sequence PY*LICK (where Y*
represents phosphotyrosine), and a polypeptide having the sequence Y*LPQTV
(where Y* represents phosphotyrosine). Additional non-limiting examples of
STAT3
inhibitors are described in Yue and Turkson Expert Opin Investig Drugs. 2009
Jan;
18(1): 45-56, the entire content of which is incorporated herein by reference.
Other STAT3 inhibitors include: El: 4_-Bromo-phenyl -2-N- aminoacyl -1 1-
dioxide-
benzo [b] thiophene; E2: 4_-bromo -2-N- (4- fluorophenyl) alany1-1,1-dioxide,
benzo
[b] thiophene; E3: 4_-bromo - benzo 2-N- (4-methoxyphenyl) alany1-1,1-dioxide
[b]
thiophene; E4: 4_-bromo -2-N- aminoacyl-p-tolyI-1,1- oxidation benzo [b]
thiophene;
E5: 4_-bromo -2-N- (4- chlorophenyl) alany1-1,1-dioxide, benzo [b] thiophene;
E6: 4_-
bromo -2-N - benzo (3-chlorophenyl) alany1-111-dioxide [b] thiophene; E7: 4_-
bromo -
2-N- (2- chlorophenyl) alany1-1,1-dioxide benzo [b] thiophene; E8: 4_-bromo -2-
N- (3-
chloro-4-fluorophenyl) alany1-1,1-dioxide, benzo [b] thiophene; E9: 4_-chloro -
2-N-
aminoacyl-pheny1-1,1-dioxide, benzo [b] thiophene; El 0: 5_-bromo-phenyl -2-N-
aminoacy1-1,1-dioxide, benzo [b] thiophene; Ell: 6_ bromo-phenyl -2-N-
aminoacyl-
1,1-dioxide, benzo [b] thiophene; El 2: 2-N- aminoacyl-phenyl-1,1-dioxide,
benzo [b]
thiophene; E13: 5_-nitro-phenyl -2-N- Acy1-1,1-dioxide, benzo [b] thiophene;
El 4: 5_-
bromo-n-butyl -2-N- aminoacy1-1,1-dioxide, benzo [b] thiophene; E15: 5_ bromo -
2-
N- am inoacyl-t-buty1-1,1-dioxide, benzo [b] thiophene; El 6: 5_-bromo -2-N-
isopropyl-
alany1-1,1-benzo [b] dioxide thiophene; El 7: 5_-bromo -2-N- cyclohexyl-alany1-
1,1-
benzo [b] thiophene dioxide; El 8: 5_-bromo -2-N- [(3s, 5s, 7s) -I -
adamantyl] -1,1-
aminoacyl dioxide benzo [b] thiophene; El 9: 4_-bromo-benzo -2-N- benzyl-
aminoacyl-
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
1,1-dioxide [b] thiophene; E20: 4_-bromo -2-N- (4- bromophenethyl) benzo-1,1-
dioxide aminoacyl [b] thiophene; E21: 5_-bromo -2-N- (4- phenoxy- phenyl)
amino-
benzo-1,1-dioxide group [b] thiophene; E22: 5_-bromo -2-N- [4- (1- piperidinyl-
carbonyl) phenyl] -1,1-am inoacyl dioxide benzo [b] thiophene; E23: 5_-bromo -
2-N-
[4- (4- morpholin-ylcarbonyl) phenyl] carboxamido-1,1-dioxide, benzo [b]
thiophene;
E24: 5_-bromo -2-N- [4- (N- methyl -N- phenyl) carbamoyl] phenyl-carboxamido-
1,1-
dioxide, benzo [b] thiophene; E25: 4 _ bromo-2-p-tolyl-carboxy-1,1-dioxide [b]
thiophene; E26: 5_ bromo -2-N, N- diethyl-1 A -dioxide am inoacyl
benzothienyl; E27:
5_-bromo -2- (1- pyrroly1) carbonyl-1,1-dioxide, benzo [b] thiophene; E28: 5_-
bromo-2-
(I-piperidyl) carbonyl-1,1-dioxide, benzo [b] thiophene; E29: 5_-bromo-2- (2-
methyl-
piperidine yl) carbonyl-1,1-dioxide, benzo [b] thiophene; E30: benzo 5_-bromo-
2- (3-
methy1-1-piperidinyl) carbonyl-IA-dioxide [b] thiophene; E31: 5_-bronno-2-
morpholino-carbony1-1,1-dioxide, benzo [b] thiophene; E32: 5_-bromo-2- (4-
ethy1-1-
piperazinyl) carbonyl-1 , 1- dioxide-benzo [b] thiophene; E33: 5_-bromo -2- (N-
methyl
-N- phenyl) benzo-1,1-dioxide aminoacyl [b] thiophene; E34: 4 _ bromo -2- (1-
piperidinyl) carbonyl-I A-dioxide, benzo [b] thiophene; E35: 5_
trifluoromethyl -2- (1-
piperidinyl) carbonyl-1,1 - dioxide benzo [b] thiophene; E36: 4_-bromo-2-
methoxycarbony1-1,1-dioxide, benzo [b] thiophene; E37: 2_ methoxycarbony1-1,1-
oxidation benzo [b] thiophene; E38: benzo 5_ acetamido -2-N- phenyl-1, -
dioxide
aminoacyl [b] thiophene; E39: 5_ benzoylamino -2-N - aminoacyl phenyl-1 ,I-
dioxide,
benzo [b] thiophene; E40: 5_ of Methylbenzamido -2-N- aminoacyl-phenyl-1,1-
dioxide, benzo [b] thiophene; E41: 5_ Trifluoromethyl-benzoyl-phenylcarbamoyl
group
-2-N- acyl-1,1 -dioxide, benzo [b] thiophene; E42: 5_ p-chlorobenzoyl-N-
phenylcarbamoyl group an acyl -2-1,1-benzo [b] thiophene dioxide; E43: 5_-
cyclohexyl-carboxamido -2-N- phenyl-aminoacyl-1,1-dioxide, benzo [b]
thiophene; or
E44: 5_ benzamido -2- (1- piperidinyl) carbonyl 1,1-dioxide, benzo [b]
thiophene.
e. GSK3 Inhibitors
In certain embodiments, the anti-PESC agent is an inhibitor of glycogen
synthase
kinase 3 (GSK-3), also referred to here as a GSK-3 inhibitor. In certain
embodiments,
the GSK-3 inhibitor can be an inhibitor of both GSK-3a and GSK-3I3. In certain
embodiments, the GSK-3 inhibitor is a selective inhibitor of GSK-3a relative
to GSK-
91
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
30. In certain embodiments, the GSK-3 inhibitor is a selective inhibitor of
GSK-313
relative to GSK-3cc.
G8K3 inhibitor such as AZD1080. AZD1080 is a selective, orally active, brain
permeable GSK3 inhibitor, inhibits human GSK3a and GSK3 I3 with Ki of 6_9 nM
and
31 nM, respectively, shows >14-fold selectivity against CDK2, CDK5, CDK1 and
Erk2.
Other examples of inhibitors of GSK-3 include aloisines (such as aloisine A
and
aloisine B), hymenialdisine (such as dibromohymenialdisine), indirubins (such
as 5,5'-
dibromo- indirubin), maleim ides, in particular macrocyclic
bisindolylmaleimides (such
as Ro 31 -8220, SB-216763, SB-415286, or 3F8), and muscarinic agonists (such
as
AF102B and AFI50).
In certain embodiments, the GSK3 inhibitor can be 6-bronnoindirubin-3'-oxinne
(B10),
CHIR-99021, SB216763, CH IR-98014, TVVS119, IM-12, 1-Azakenpaullone, AR-
A014418, SB415286, AZD1080, AZD2858, indirubin, A 1070722, TCS 2002,
Tideglusib, or any derivatives thereof.
In another embodiment, the GSK-3 inhibitor comprises flavopiridol,
kenpaullone,
alsterpaullone, azakenpaullone, pyrazolopyridine, CHIR98014, CHIR99021 , CHIR-
637, CT20026, SU9516, ARA014418, and staurosporine.
Exemplary GSK3 inhibitor can be selected from the group consisting of Lit,
(27,3'E)-
6-bromoindirubin-3'-oxime (B10), (2Z,31EJ-6-bromoindirubin-3'-acetoxime (B10-
acet-
oxime), SB-216763 (3-(2,4-dichlorophenyI)-4-(1 -methyl-1 H-indo1-3-y1)-1 H-
pyrrole-
2,5- dione), SB-415286 (3-[(3-chloro-4-hydroxyphenyl)amino]-4-(2-nitropheny1)-
1 H-
pyrrol-2,5- dione), enzastaurin (3-(1 -methylindo1-3-y1)-4-[1 -[1 -(pyridin-2-
ylmethyl)piperidin-4-yl]indol- 3-yl]pyrrole-2,5-dione), L803-mts (Myr-N-Gly-
Lys-Glu-
Ala-Pro-Pro-Ala-Pro-Pro-Gln- pSer(P03H)-Pro-NH2), NP031 1 12 (4-benzy1-2-
naphtalen-1 -y1-1 ,2,4-thiadiazolidine-3,5- dione), paliperidone palmitate (3-
(2-(4-(6-
fluoro-1 ,2-benzisoxazol-3-y1)-1 -piperidinypethyl)- 6,7,8,9-tetrahydro-2-
methy1-4-oxo-
4H-pyrido(1 ,2-a)pyrimidin-9-y1 ester), valproic acid (2- propylpentanoic
acid), TDZD-
8 (4-benzy1-2-methy1-1 ,2,4-thiadiazolidine-3,5-dione), and 9-
hydroxyrisperidone
(6,7,8,9-tetrahydro-3-(2-(4-(6-fluoro-1 ,2-benzisoxazol-3-y1)-1 -
piperidinypethyl)-9-
hydroxy-2-methyl-4H-pyrido[2, 1 -a]pyrim idin-4-one).
Among GSK-3 inhibitors, derivatives of the bis-indole indirubin (collectively
referred to
as indirubins) appear as a class of original and promising tools and agents.
Their
92
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
moderate selectivity might be inconvenient when used as a research reagent,
but their
combined effects on several disease-relevant targets (in particular CDKs and
GSK-3)
may constitute an advantage for potential therapeutic applications. Among many
indirubins, 6-bromo- indirubin-3'-oxime (B10) has been widely used to
investigate the
physiological role of GSK-3 in various cellular settings and to alter the fate
of
embryonic stem cells.
In one embodiment, the GS K-3 inhibitor is a compound of formula:
X1 /
,--3---00n
1
r-----3
Min H _
-------1:5-----N
NN ,
'RIIR-
X2
wherein X1 and X2, independently of each other, are 0, S, N-OR3, N(Z1), or two
groups
independently selected from H, F, CI, Br, 1, NO2, phenyl, and (CrC6)alkyl, and
wherein
R3 is hydrogen, (C C6)alkyl, or (C C6)alkyl-C(0)-;
each Y, independently of each other, is H, (C C6)alkyl, (C C6)alkyl-C(0)-, (C
C6)alkyl-
C(0)0-, phenyl, N(Z1)(Z2), sulfonyl, phosphonyl, F, CI, Br, or 1;
Z1 and Z2, independently of each other, are H, (d-C6)alkyl, phenyl, benzyl, or
Z1 and
Z2 together with the nitrogen to which they are attached represent a 5, 6, or
7-
membered heterocyclyl;
n and m, independently of each other, are 0, 1 , 2, 3, or 4;
R1 and R2, independently of each other, are H, (CrC6)alkyl, (CrC6)alkyl-C(0)-,
phenyl,
benzyl, or benzoyl;
and wherein alkyi is branched or straight-chain alkyi, optionally substituted
with 1 , 2,
3, 4, or 5 OH, N(Z1)(Z2), (CrC6)alkyl, phenyl, benzyl, F, CI, Br, or 1; and
wherein any
phenyl, benzyl, or benzoyl is optionally substituted with 1, 2, 3, 4, or 5 OH,
N(Z1)(Z2),
(C C6)alkyl, F, CI, Br, or I;
or a salt thereof.
In one embodiment? X1 is 0 and X2 is N-OH, or X1 is N-OH and X2is 0. In
another
embodiment, one Y is Br. In another embodiment, one Y is Br at the 6'-
position. In
93
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
another embodiment, n is 0 and m is 1 , or n is 1 and m is 0. In another
embodiment
R1 and R2 are
H.
In one embodiment, the GSK-3 inhibitor comprises 6-bromoindirubin-3'-oxime of
formula
H 0, Br
N
/
_
N N H
H 0
or a salt thereof.
In one embodiment, the GSK-3 inhibitor comprises 6-bromoindirubin-3'-acetoxime
of
formula
7-
0,
Br
N
/
-
N H
N
H 10 0
or a salt thereof.
I HSP90, HSP70 and dual HSP90170 Inhibitors
In certain embodiments, the anti-PESC agent is an HSP90 Inhibitor or an HSP70
inhibitor or both. An exemplary anti-PESC agent is Nanchangmycin
94
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
H3y 11,.ic
'O F4 3
H3
HS Ira H s
11.3CliE IN , , f.- pCbt %%Ica A '4013
..,..,..õ.õ(Scr
0 0 e A b-F v 4 0.1.ntik,
0 Ft Ft3 HO OH
>h ill
c CH3
94A cH4 ch3
,?;.. :.
HO CH3 Nanchangnwcin
Other examples of Hsp90 inhibitors include, but are not limited to,
geldanamycin,
rad icicol, 17-N-Allylamino-17-demethoxygeldanamycin/tanespicmycin/17AAG (BM
S),
herbimycin A, novobiocin sodium (U-6591), 17-GMB-APA-GA, macbecin I, CCT
018159, gedunin, PU24FC1, PU-H71, PU-DZ8, PU3, AUY922 (Novartis), HSP990
(Novartis), retaspimycin hydrochloride/IP1-504 (Infinity), B1113021/CNF2024
(Biogen
!deo), STA-9090 (Synta), SNX-5422/mesylate (Pfizer), 131113028 (Biogen !dee),
KW-
2478 (Kyowa Hakko Kirin), AT13387 (Astex), XL888 (Exelixis), MPC-3100
(Myriad),
ABI-010/nab (nanoparticle, albumin bound)-17AAG (Abraxis), 17-
am inodem ethoxygeldanam ycin (IPI-493),
17-dimethylaminoethylam ino-17-
demethoxygeldanamycin (17-DMAG), XL888, SNX-2112, SNX-7081, ganetespib
(STA-9090), AUY922, Debio0932, BI113028, BIIB021, MPC-3100, MPC-0767, PU3,
PU-H58, DS-2248, KW-2478, CCT018159, CCT0129397, BJ-B11, elesclomol (STA-
4783), G3130, gedunin, herbimycin, radester, KNK437, HSP990, or NVP-BEP800.
q
WO, .,....k.k.0 0 0
1 t jt, me
4---- in: -sr
i
0
---' ,
1 3 ' ' ki
H
I
Me'
H3C0 N
0.3
CH3
--_,
.3c0
0 CON H2
getclarativerin H erbbnyein A
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
IV. Combination Therapies - ESO Regenerative agent
In certain embodiments, the anti-P ESC agent can be administered conjointly
with one
or more agents that selectively promote proliferation or other regenerative
and wound
healing activities of normal epithelial stem cells. Conjoint administration of
these "ESO
Regenerative agents" may be accomplished by administration of a single co-
formulation, by simultaneous administration or by administration at separate
times.
In certain embodiments, the anti-P ESC agent can be administered conjointly
with one
or more agents that selectively promote proliferation or other regenerative
and wound
healing activities of normal esophageal stem cells. Conjoint administration of
these
"esophageal ESO Regenerative agents" may be accomplished by administration of
a
single co-formulation, by simultaneous administration or by administration at
separate
times.
a. ABL kinase inhibitor
In certain embodiments, the ESO Regenerative agent is pan-inhibitor of ABL
kinase
inhibitor, preferably a BCR-ABL kinase inhibitor Exemplary pan-inhibitor
include
imatinib, nilotinib, dasatinib, bosutinib and ponatinib, and is preferably
ponatinib.
b. FLT3 Inhibitors
In certain embodiments, the ESO Regenerative agent is a FLT3 inhibitor.
Exemplary
FLT3 inhibitors to be used herein are quizartinib (AC220), crenolanib (CP-
868596),
midostaurin (PKC-412), lestaurtinib (CEP-701), 4SC-203, TTT-3002, sorafenib
(Bay-
43-0006), Ponatinib (AP-24534), sunitinib (SU-11248), and/or tandutinib (MLN-
0518),
or (a) pharmaceutically acceptable salt(s), solvate(s), and/or hydrate(s)
thereof.
Preferably, the FMS-like tyrosine kinase 3 (FLT3) inhibitor is quizartinib
(AC220) or
pharmaceutically acceptable salt(s), solvate(s), and/or hydrate(s) thereof.
These and further exemplary inhibitors to be used herein are described in more
detail
below.
Brand Name: Quizartinib
Structure:
96
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
S N
N
0 N
rr1/2.. N jr..44%ir-%=ee
N N etiejs<
0
Affinities: FLT3 (1.6 nM), KIT (4.8 nM), PDGFRB (7.7 nM), RET (9.9 nM), PDGFRA
(11 nM),CSF1R (12 nM)
Brand Name: Crenolanib
Structure:
====1/2.%
I ee,
N N N
0
Ii
N
1Js;
?coTi
0
Affinities: FLT3, PDGFRb
Brand Name: Midostaurin
Structure:
97
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
o N
=
14 +dr
N.). 0 si-Ary
0 RI
11011
Affinities: PKNI (9.3 nM), TBKI (9.3 nM), FLT3 (11 nM), JAK3 (12 nM), MLKI (15
nM),
and
30 targets in the range 15-110 nM
Brand Name: Lestaurtinib
N
0
INNS
0
0 ______________________________________________________________________
0
Affinities: FLT3, TRKA, TRKB, TRKC
Brand Name: 4SC-203
Structure:
98
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
...-
N
0
ii ,_ N N gia
N S
0 la
Affinities: FLT3, VEGFR
Structure:
li
N 0
171-4002
a
tit
14
0
Me4 l0,21 i...ii
H2N S
0,0;45% WPM
H
Affinities: FLT3 (Wall, Blood (ASH Annual Meeting Abstracts). 2012; 120:866);
LRRIQ (Yao, Human molecular genetics. 2013;22(2):328-44).
Clinical Phase: Preclinical
Developer: Tautatis (originator)
Brand Name: Sorafenib
Code Name: Bay-43-0006
Structure:
99
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
0 crekie.õ
0
11101 NA.N 101
N
1UPAC Name:
444-[344-Chloro-3-
(trifluoromethypphenyliureido]phenoxy]-N-
methylpyridine-2- carboxamide
Affinities: DDRI (1.5 nM), HIPK4 (3 nM), ZAK (6 nM), DDR2 (7 nM), FLT3 (13
nM), and
15 targets in the range 13-130 nM (Zarrinkar, Gunawardane et at 2009, loc.
cit.)
Clinical Phase: Launched (renal and heptacellular carcinoma), Phase 110 (blood
cancer) Developer: Bayer
Brand Name: Ponatinib
Code Name: AP-24534 Structure:
73
SOD
Sill 0
N
Nor
I U PAC Name:
3-[2-(Imidazo[1,2-b]pyridazin-
3-ypethynyl]-4-methyl-N44-(4-
methylpiperazin-1- ylmethyl)-3-(trifluoromethyl)phenylibenzamide
Affinities: BCR-ABL, FLT3, KIT, FGFR1, PDGFRa (Gozgit, Mol Cancer Ther.
2011;10(6):1028-35).
Clinical Phase: Phase II (AML)
Developer: Ariad Pharmaceuticals (originator)
Brand Name: Sunitinib
Code Name: SU-11248
Structure:
100
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
.
r
...,...N......"... N ...,.......0e-
N
/ X
I N
41
1 0
N
0
0 nenc
i
IU PAC Name: (Z)-N-[2-( D iethylann ino)ethyI]-5-(5-fluoro-2-oxo-2 , 3-d
ihydro-IH-indo1-3-
ylidenemethyl)-2,4-dimethyl-IH-pyrrole-3-carboxam ide 2(S)-hydroxybutanedioic
acid
(1 :1) 1412-(Diethylamino)ethy1]-5-[(Z)-(5-fluoro-2-oxo-I,2-dihydro-3H-indol-
3 -
ylidene)methyl] -2,4-dimethyl- 1 H -pyrrole-3 -carboxamide L-malate
Affinities: PDGFRB (0.075 nM), KIT (0_37 nM), FLT3 (0.47 nM), PDGFRA (0_79
nM),
DRAK1 (1.0 nM), VEGFR2 (1.5 nM), FLT1 (1.8 nM), CSF1R (2.0 nM) (Zarrinkar,
Gunawardane et at 2009, loc. cit.)
Clinical Phase: Launched (renal cell carcinoma, gastrointestinal stromal
cancer,
neuroendocrine pancreas), phase I (AML)
Developer: Pfizer (Originator)
Brand Name: Tandutinib
Code Name: MLN-0518
Structure:
0
N
N 101 1,....õ
C )
0.
N
-Piarn S `"1/4- 11
0 .e."'w-A"el.e.'N' 0 1111111r Iteli
101
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
I UP AC Name: N-(4-
lsopropoxypheny1)-4[6-methoxy-7[3-(l -
piperidinyl)propoxy]quinazolin-4- ylipiperazine- 1 -carboxamide
Affinities: PDGFRA (2.4nM), KIT (2.7nM), FLT3 (3nM), PDGFRB (4.5nM), CSF1R
(4.9nM) (Zarrinkar, Gunawardane et at. 2009, loc. cit.)
Clinical Phase: discontinued
Developer: Kyowa Hakko Kirin (Originator), Millennium Pharmaceuticals
(Originator),
Code Name: FF-10101
Structure:
As 0
NH
õ, l
---
N N
H
National Cancer Institute, Takeda (Originator) FLT3 inhibitors to be used in
accordance with the present disclosure are not limited to the herein described
or
further known exemplary inhibitors. Accordingly, also further inhibitors or
even yet
unknown inhibitors may be used in accordance with the present disclosure. Such
inhibitors may be identified by the methods described and provided herein and
methods known in the art, like high- throughput screening using biochemical
assays
for inhibition of FLT3.
Assays for screening potential FLT3 inhibitors and, in particular, for
identifying FLT3
inhibitors as defined herein, comprise, for example, in vitro competition
binding assays
to quantitatively measure interactions between test compounds and
recombinantly
expressed kinases1 (Fabian et al; Nat Blotechnol. 2005 23(3):329-36). Hereby,
competition with immobilized capture compounds and free test compounds is
performed. Test compounds that bind the kinase active site will reduce the
amount of
kinase captured on solid support, whereas test molecules that do not bind the
kinase
have no effect on the amount of kinase captured on the solid support.
Furthermore,
inhibitor selectivity can also be assessed in parallel enzymatic assays for a
set of
102
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
recombinant protein kinases.2.3 (Davies et at, Biochem. J. 2000 35(1): 95-105;
Bain
et at Biochem. J. 2003 37(1): 199-204). These assays are based on the
measurement
of the inhibitory effect of a kinase inhibitor and determine the concentration
of
compound required for 50% inhibition of the protein kinases of interest.
Proteomics
methods are also an efficient tool to identify cellular targets of kinase
inhibitors.
Kinases are enriched from cellular lysates by immobilized capture compounds,
so the
native target spectrum of a kinase inhibitor can be determined.4 (Godl et al;.
Proc Nati
Acad Sci USA. 2003 100(26): 5434-9).
Assays for screening of potential inhibitors and, in particular, for
identifying inhibitors
as defined herein, are, for example, described in the following papers:
= Fabian et aL, Nat Biotechnot 2005 23(3):329-36
= Davies et aL, Biochem. J 2000 351 : 95-105.
= Bain et al., Biochem. J. 2003 371 : 199-204.
= Godl et al., Proc Nat! Acad Sci USA. 2003 100(26): 15434-9.
The above papers are incorporated herein in their entirety by reference.
V. Combination Therapies ¨ Other agents
In certain embodiments, the anti-P ESC agent can be administered conjointly
with one
or more agents that have other beneficial local activities in esophagus.
Illustrative
categories and specific examples of active drugs include: (a) antitussives,
such as
dextromethorphan, dextromethorphan hydrobromide, noscapine, carbetapentane
citrate, and chlophedianol hydrochloride; (b) antihistamines, such as
chlorpheniramine
maleate, phenindamine tartrate, pyrilamine maleate, doxylamine succinate, and
phenyltoloxannine citrate; (c) antipyretics and analgesics such as
acetaminophen,
aspirin and ibuprofen; (d) antacids such as aluminum hydroxide and magnesium
hydroxide, (e) anti-infective agents such as antifungals, antivirals,
antiseptics and
antibiotics, (t) chemotherapeutic agents.
VI. Exemplary Formulations
In certain embodiments, the anti-PESC agents is formulated for topical
administration
as part of a bioadhesive formulation. Bioadhesive polymers have extensively
been
103
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
employed in transmucosal drug delivery systems and can be readily adapted for
use
in delivery of the subject anti-PESC agents to the esophagus, particularly the
areas of
lesions and tumor growth. In general terms, adhesion of polymers to tissues
may be
achieved by (i) physical or mechanical bonds, (ii) primary or covalent
chemical bonds,
and/or (iii) secondary chemical bonds (i.e., ionic). Physical or mechanical
bonds can
result from deposition and inclusion of the adhesive material in the crevices
of the
mucus or the folds of the mucosa_ Secondary chemical bonds, contributing to
bioadhesive properties, consist of dispersive interactions (i.e.. Van der
Waals
interactions) and stronger specific interactions, which include hydrogen
bonds. The
hydrophilic functional groups responsible for forming hydrogen bonds are the
hydroxyl
(-OH) and the carboxylic groups (¨COOH). When these materials are incorporated
into pharmaceutical formulations, drug absorption by mucosal cells may be
enhanced
and/or the drug may be released at the site for an extended period of time.
Merely to
illustrate, the bioadhesive can be a hydrophilic polymer, a hydrogel, a co-
polymers/interpolymer complex or a thiolated polymer.
= Hydrophilic polymers: These are water-soluble polymers that swell when
they
come in contact with water and eventually undergo complete dissolution.
Systems coated with these polymers show high bioadhesiveness to the mucosa
in dry state but the bioadhesive nature deteriorates as they start dissolving_
As
a result, their bioadhesiveness is short-lived. An example is poly (acrylic
acid).
= Hydrogels: These are three-dimensional polymer networks of hydrophilic
polymers which are cross-linked either by chemical or physical bonds. These
polymers swell when they come in contact with water. The extent of swelling
depends upon the degree of crosslinking. Examples are polycarbophil,
carbopol and polyox.
= Co-polynners/Interpolyrner complex: A block copolymer is formed when the
reaction is carried out in a stepwise manner, leading to a structure with long
sequences or blocks of one monomer alternating with long sequences of the
other. There are also graft copolymers, in which entire chains of one kind
(e.g.,
polystyrene) are made to grow out of the sides of chains of another kind
(e.g.,
polybutadiene), resulting in a product that is less brittle and more impact-
resistant Hydrogen bonding is a major driving force for interpolymer
interactions.
104
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
= Thiolated polymers (Thionners): These are hydrophilic macromolecules
exhibiting free thiol groups on the polymeric backbone. Based on
thiol/disulfide
exchange reactions and/or a simple oxidation process disulfide bonds are
formed between such polymers and cysteine-rich subdomains of mucus
glycoproteins building up the mucus gel layer. So far, the cationic thiorners,
chitosan¨cysteine, chitosan¨thiobutylamidine as well as chitosan¨thioglycolic
acid, and the anionic thiomers, poly (acylic acid)¨cysteine, poly (acrylic
acid)¨
cysteamine, carboxymethylcellulose¨cysteine and alginate¨ cysteine, have
been generated. Due to the immobilisation of thiol groups on mucoadhesive
basis polymers, their mucoadhesive properties are 2- up to 140- fold improved.
In certain embodiments, the bioadhesive polymer can be selected from
poly(acrylic
acid), tragacanth, poly(methylvinylether comaleic anhydride), poly(ethylene
oxide),
methyl-cellulose, sodium alginate, hydroxypropylmethylcellulose, karaya gum,
methylethyl cellulose (and cellulose derivatives such as Metolose), soluble
starch,
gelatin, pectin, poly(vinyl pyrrolidone), poly(ethylene glycol), poly(vinyl
alcohol),
poly( hydroxyethyl-m ethacry late),
hydroxypropylcellulose, sodium
carboxymethylcellulose or chitosan.
Other suitable bioadhesive polymers are described in U.S. Pat. No. 6,235,313
to
Mathiowitz et at, the teachings of which are incorporated herein by reference,
and
include polyhydroxy acids, such as poly(lactic acid), polystyrene,
polyhyaluronic acids,
casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan;
polyacrylates, such as poly(methyl methacrylates), poly(ethyl methacrylates),
poly
butylmethacrylate), poly-(isobutylnnethacrylate), poly(hexlmethacrylate),
poly(isodecl
rnethacrylate), poly(lauryl methacrylate), poly(phenyl nnethacrylate),
poly(methyl
acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), and
poly(octadecl acrylate);
polyacrylamides; poly(fumaric-co-sebacic)acid, poly(bis carboxy phenoxy
propane-
co-sebacic anhydride), polyorthoesters, and copolymers, blends and mixtures
thereof.
In certain embodiments, the bioadhesive is an alginate. Alginic acid and its
salts
associates with sodium and potassium bicarbonate have shown that, after
entering a
more acidic environment they form a viscous suspension (or a gel) exerting
protecting
activity over gastric mucosa. These properties are readily adaptable for
topical delivery
to the esophagus, particularly the lower esophagus. The scientific and patent
literature
on its activity is wide. Thus, for example, for delivery to the esophagus:
Mandel K. G.;
105
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Daggy B. P.; Brodie D. A; Jacoby, H. L., 2000. Review article: Alginate-raft
formulations in the treatment of heartburn and acid reflux. Aliment Pharmacol
Ther.
14 669-690, which is incorporated by reference herein in its entirety; and
Bioadhesive
esophageal bandages: protection against acid and pepsin injury. Man Tang,
Peter
Dettmar, Hannah Batchelor¨/nternationa/ Journal of Pharmaceutics 292 (2005)-
169-177, which is incorporated by reference herein in its entirety.
In certain embodiments, the bioadhesive is a bioadhesive hydrogel. Bioadhesive
hydrogels are well known in art and suitable hydrogels that be used for
delivery of the
anti-P ESC agents of the present disclosure are described in a wide range of
scientific
and patent literature on its activity is wide. An exemplary hydrogel
formulation is
described in Co!laud et aL "Clinical evaluation of bioadhesive hydrogels for
topical
delivery of hexylaminolevulinale to Barrett's esophagus" J Control Release.
2007 Nov
20; 123(3):203-10.
a. Bioadhesive Microparticle formulations
In certain embodiments, the anti-PESC agent (optionally with other active
agents) are
formulated into adhesive polymeric microspheres have been selected on the
basis of
the physical and chemical bonds formed as a function of chemical composition
and
physical characteristics, such as surface area, as described in detail below.
These
microspheres are characterized by adhesive forces to mucosa of greater than 11
mN/cm2 on esophageal tissue. The size of these microspheres can range from
between a nanoparticle to a millimeter in diameter. The adhesive force is a
function of
polymer composition, biological substrate, particle morphology, particle
geometry
(e.g., diameter) and surface modification.
Suitable polymers that can be used to form bioadhesive microspheres include
soluble
and insoluble, biodegradable and nonbiodegradable polymers. These can be
hydrogels or thermoplastics, homopolymers, copolymers or blends, natural or
synthetic. The preferred polymers are synthetic polymers, with controlled
synthesis
and degradation characteristics. Most preferred polymers are copolymers of
fumaric
acid and sebacic acid, which have unusually good bioadhesive properties when
administered to the gastrointestinal.
In the past, two classes of polymers have appeared to show useful bioadhesive
properties: hydrophilic polymers and hydrogels. In the large class of
hydrophilic
106
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
polymers, those containing carboxylic groups (e.g., poly[acrylic acid])
exhibit the best
bioadhesive properties. One could infer that polymers with the highest
concentrations
of carboxylic groups should be the materials of choice for bioadhesion on soft
tissues.
In other studies, the most promising polymers were sodium alginate,
carboxymethylcellulose, hydroxymethylcellulose and methylcellulose. Some of
these
materials are water-soluble, while others are hydrogels.
Rapidly bioerodible polymers such as poly[lactide-co-glycolide],
polyanhydrides, and
polyorthoesters, whose carboxylic groups are exposed on the external surface
as their
smooth surface erodes, are excellent candidates for bioadhesive drug delivery
systems. In addition, polymers containing labile bonds, such as polyanhydrides
and
polyesters, are well known for their hydrolytic reactivity. Their hydrolytic
degradation
rates can generally be altered by simple changes in the polymer backbone.
Representative natural polymers include proteins, such as zein, modified zein,
casein,
gelatin, gluten, serum albumin, or collagen, and polysaccharides, such as
cellulose,
dextrans, polyhyaluronic acid, polymers of acrylic and methacrylic esters and
alginic
acid. These are not preferred due to higher levels of variability in the
characteristics of
the final products, as well as in degradation following administration.
Synthetically
modified natural polymers include alkyl celluloses, hydroxyalkyl celluloses,
cellulose
ethers, cellulose esters, and nitrocelluloses.
Representative synthetic polymers include polyphosphazines, poly(vinyl
alcohols),
polyamides, polycarbonates, polyalkylenes, polyacrylamides, polyalkylene
glycols,
polyalkylene oxides, polyalkylene terephthalates, polyvinyl ethers, polyvinyl
esters,
polyvinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes,
polyurethanes
and copolymers thereof. Other polymers of interest include, but are not
limited to,
methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl
methyl
cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose
propionate,
cellulose acetate butyrate, cellulose acetate phthalate, carboxynnethyl
cellulose,
cellulose triacetate, cellulose sulfate sodium salt, poly(methyl
methacrylate), poly(ethyl
methacrylate), poly(butyl methacrylate), poly(isobutyl methacrylate),
poly(hexyl
methacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate),
poly(phenyl
methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl
acrylate),
poly(octadecyl acrylate) polyethylene, polypropylene, poly(ethylene glycol),
poly(ethylene oxide), poly (ethylene terephthalate), poly(vinyl acetate),
polyvinyl
107
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
chloride, polystyrene, polyvinyl pyrrolidone, and polyvinylphenol.
Representative
bioerodible polymers include polylactides, polyglycolides and copolymers
thereof,
poly(ethylene terephthalate), poly(butic acid), poly(valeric acid),
poly(lactide-co-
caprolactone), poly[lactide-co-glycolide], polyanhydrides, polyorthoesters,
blends and
copolymers thereof.
These polymers can be obtained from sources such as Sigma Chemical Co., St.
Louis,
Mo., Polysciences, Warrenton, Pa., Aldrich, Milwaukee, Wis., Fluka,
Ronkonkoma,
N.Y., and BioRad, Richmond, Calif. or else synthesized from monomers obtained
from
these suppliers using standard techniques.
In some instances, the polymeric material could be modified to improve
bioadhesion
either before or after the fabrication of microspheres. For example, the
polymers can
be modified by increasing the number of carboxylic groups accessible during
biodegradation, or on the polymer surface. The polymers can also be modified
by
binding amino groups to the polymer. The polymers can also be modified using
any of
a number of different coupling chemistries that covalently attach ligand
molecules with
bioadhesive properties to the surface-exposed molecules of the polymeric
microspheres.
One useful protocol involves the "activation" of hydroxyl groups on polymer
chains
with the agent, carbonyldiimidazole (COI) in aprotic solvents such as DMSO,
acetone,
or THF. CDI forms an imidazolyl carbamate complex with the hydroxyl group
which
may be displaced by binding the free amino group of a ligand such as a
protein. The
reaction is an N-nucleophilic substitution and results in a stable N-
alkylcarbamate
linkage of the ligand to the polymer. The "coupling" of the ligand to the
"activated"
polymer matrix is maximal in the pH range of 9-10 and normally requires at
least 24
28 hrs. The resulting ligand-polymer complex is stable and resists
hydrolysis for extended
periods of time.
Another coupling method involves the use of 1-ethyl-3-(3-dimethylaminopropyl)
carbodiimide (EDAC) or "water-soluble CDI" in conjunction with N-
hydroxylsulfosuccinimide (sulfo NHS) to couple the exposed carboxylic groups
of
polymers to the free amino groups of ligands in a totally aqueous environment
at the
physiological pH of 7Ø Briefly, EDAC and sulfo-NHS form an activated ester
with the
carboxylic acid groups of the polymer which react with the amine end of a
ligand to
108
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
form a peptide bond. The resulting peptide bond is resistant to hydrolysis.
The use of
sulfo-NHS in the reaction increases the efficiency of the EDAC coupling by a
factor of
ten-fold and provides for exceptionally gentle conditions that ensure the
viability of the
I igand-polymer complex.
By using either of these protocols it is possible to "activate" almost all
polymers
containing either hydroxyl or carboxyl groups in a suitable solvent system
that will not
dissolve the polymer matrix_
A useful coupling procedure for attaching ligands with free hydroxyl and
carboxyl
groups to polymers involves the use of the cross-linking agent,
divinylsulfone. This
method would be useful for attaching sugars or other hydroxylic compounds with
bioadhesive properties to hydroxylic matrices. Briefly, the activation
involves the
reaction of divinylsulfone to the hydroxyl groups of the polymer, forming the
vinylsulfonyl ethyl ether of the polymer. The vinyl groups will couple to
alcohols,
phenols and even amines. Activation and coupling take place at pH 11. The
linkage is
stable in the pH range from 1-8 and is suitable for transit through the
intestine.
Any suitable coupling method known to those skilled in the art for the
coupling of
ligands and polymers with double bonds, including the use of UV crosslinking,
may be
used for attachment of bioadhesive ligands to the polymeric microspheres
described
herein. Any polymer that can be modified through the attachment of lectins can
be
used as a bioadhesive polymer for purposes of drug delivery or imaging.
Lectins that can be covalently attached to microspheres to render them target
specific
to the mucin and mucosal cell layer could be used as bioadhesives. Useful
lectin
ligands include lectins isolated from: Abrus precatroius, Agaricus bisporus,
Anguilla
anguilla, Arachis hypogaea, Pandeiraea simplicifolia, Bauhinia purpurea,
Caragan
arobrescens, Cicer arietinum, Codiurn fragile, Datura stramonium, Dolichos
biflorus,
Erythrina corallodendron, Erythrina cristagalli, Euonymus europaeus, Glycine
max,
Helix aspersa, Helix pomatia, Lathyrus odoratus, Lens culinaris, Limulus
polyphemus,
Lysopersicon esculentum, Maclura pomifera, Momordica charantia, Mycoplasma
gallisepticum, Naja mocambique, as well as the lectins Concanavalin A,
Succinyl-
Concanavalin A, Triticum vulgaris, Ulex europaeus 1, II and III, Sambucus
nigra,
Maackia amurensis, Limax fluvus, Homarus americanus, Cancer antennarius, and
Lotus tetragonolobus.
109
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
The attachment of any positively charged ligand, such as polyethyleneimine or
polylysine, to any microsphere may improve bioadhesion due to the
electrostatic
attraction of the cationic groups coating the beads to the net negative charge
of the
mucus. The mucopolysaccharides and mucoproteins of the mucin layer, especially
the
sialic acid residues, are responsible for the negative charge coating. Any
ligand with
a high binding affinity for mucin could also be covalently linked to most
microspheres
with the appropriate chemistry, such as CM, and be expected to influence the
binding
of microspheres to the gut. For example, polyclonal antibodies raised against
components of mucin or else intact mucin, when covalently coupled to
microspheres,
would provide for increased bioadhesion. Similarly, antibodies directed
against
specific cell surface receptors exposed on the lumenal surface of the
intestinal tract
would increase the residence time of beads, when coupled to microspheres using
the
appropriate chemistry. The ligand affinity need not be based only on
electrostatic
charge, but other useful physical parameters such as solubility in mucin or
else specific
affinity to carbohydrate groups.
The covalent attachment of any of the natural components of mucin in either
pure or
partially purified form to the microspheres would decrease the surface tension
of the
bead-gut interface and increase the solubility of the bead in the mucin layer.
The list
of useful ligands would include but not be limited to the following: sialic
acid,
neuraminic acid, n-acetyl-neuraminic acid, n-glycolylneuraminic acid, 4-acetyl-
n-
acetylneuraminic acid, diacetyl-n-acetylneuraminic acid, glucuronic acid,
iduronic acid,
galactose, glucose, nnannose, fucose, any of the partially purified fractions
prepared
by chemical treatment of naturally occurring mucin, e.g., mucoproteins,
mucopolysaccharides and mucopolysaccharide-protein complexes, and antibodies
immunoreactive against proteins or sugar structure on the mucosa! surface.
The attachment of polyamino acids containing extra pendant carboxylic acid
side
groups, e.g., polyaspartic acid and polyglutannic acid, should also provide a
useful
means of increasing bioadhesiveness. Using polyamino acids in the 15,000 to
50,000
kDa molecular weight range would yield chains of 120 to 425 amino acid
residues
attached to the surface of the microspheres. The polyamino chains would
increase
bioadhesion by means of chain entanglement in mucin strands as well as by
increased
carboxylic charge.
110
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
As used herein, the term "microspheres" includes microparticles and
microcapsules
(having a core of a different material than the outer wall), having a diameter
in the
nanometer range up to 5 mm. The microsphere may consist entirely of
bioadhesive
polymer or have only an outer coating of bioadhesive polymer.
As characterized in the following examples, microspheres can be fabricated
from
different polymers using different methods. Polylactic acid blank microspheres
were
fabricated using three methods: solvent evaporation, as described by E.
Mathiowitz,
etal., J. Scanning Microscopy, 4, 329 (1990); L. R. Beck, etal., Fertil.
Steril., 31, 545
(1979); and S. Benita, et at, J. Pharm. Sci., 73, 1721 (1984); hot-melt
microencapsulation, as described by E. Mathiowitz, et at, Reactive Polymers,
6, 275
(1987); and spray drying_ Polyanhydrides made of bis-carboxyphenoxypropane and
sebacic acid with molar ratio of 20:80 P(CPP-SA) (20:80) (Mw 20,000) were
prepared
by hot-melt microencapsulation. Poly(fumaric-co-sebacic) (20:80) (Mw 15,000)
blank
microspheres were prepared by hot-melt microencapsulation. Polystyrene
microspheres were prepared by solvent evaporation.
In certain embodiments, the composition includes a bioadhesive matrix in which
particles (such as nanoparticles) containing the anti-PESC agents are
dispersed. In
these embodiments, the bioadhesive matrix promotes contact between the mucosa
of
the esophagus and the nanoparticles.
In certain embodiments, the drug-containing particle is a matrix, such as as a
bioerodible, bioadhesive matrix. Suitable bioerodible, bioadhesive polymers
include
bioerodible hydrogels, such as those described by Sawhney, et at, in
Macromolecules, 1993,26:581-587, the teachings of which are incorporated
herein by
reference. Representative bioerodible, bioadhesive polymers include, but are
not
limited to, synthetic polymers such as poly hydroxy acids, such as polymers of
lactic
acid and glycolic acid, polyanhydrides, poly(ortho)esters, polyesters,
polyurethanes,
poly(butic acid), poly(valeric acid), poly(caprolactone),
poly(hydroxybutyrate),
poly(lactide-co-glycolide), poly(lactide-co-caprolactone), poly(ethylene-co-
maleic
anhydride), poly(ethylene maleic anhydride-co-L-dopamine), poly(ethylene
maleic
anhydride-co-phenylalanine), poly(ethylene maleic anhydride-co-tyrosine),
poly(butadiene-co-maleic anhydride), poly(butadiene maleic anhydride-co-L-
dopamine) (pBMAD), poly(butadiene maleic anhydride-co-phenylalanine),
poly(butadiene maleic anhydride-co-tyrosine), poly(fumaric-co-
sebacic)anhydride
111
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
(P(FA:SA)), poly(bis carboxy phenoxy propane-co-sebacic anhydride) (20:80)
(poly(CCP:SA)), as well as blends comprising these polymers; and copolymers
comprising the monomers of these polymers, and natural polymers such as
alginate
and other polysaccharides, collagen, chemical derivatives thereof
(substitutions,
additions of chemical groups, for example, alkyl, alkylene, hydroxylations,
oxidations,
and other modifications routinely made by those skilled in the art), albumin
and other
hydrophilic proteins, zein and other prolamines and hydrophobic proteins,
copolymers,
blends and mixtures thereof. In general, these materials degrade either by
enzymatic
hydrolysis or exposure to water in vivo, by surface or bulk erosion.
Particles having an average particle size of between 10 nm and 10 microns are
useful
in the compositions described herein. In certain embodiments, the particles
are
nanoparticles, having a size range from about 10 nm to 1 micron, preferably
from about
10 nm to about 0.1 microns_ In particularly preferred embodiments, the
particles have
a size range from about 500 to about 600 nm. The particles can have any shape
but
are generally spherical in shape.
The compositions described herein contain a monodisperse plurality of
nanoparticles.
Preferably, the method used to form the nanoparticles produces a monodisperse
distribution of nanoparticles; however, methods producing polydisperse
nanoparticle
distributions can be used. If the method does not produce particles having a
monodisperse size distribution, the particles are separated following particle
formation
to produce a plurality of particles having the desired size range and
distribution_
Nanoparticles useful in the compositions described herein can be prepared
using any
suitable method known in the art. Common microencapsulation techniques
include,
but are not limited to, spray drying, interfacial polymerization, hot melt
encapsulation,
phase separation encapsulation (spontaneous emulsion microencapsulation,
solvent
evaporation microencapsulation, and solvent removal microencapsulation),
coacervation, low temperature microsphere formation, and phase inversion
nanoencapsulation (PIN). A brief summary of these methods is presented below.
b. Spray Drying
Methods for forming microspheres/nanospheres using spray drying techniques are
described in U.S. Pat. No. 6,620,617, to Mathiowitz et at In this method, the
polymer
is dissolved in an organic solvent such as methylene chloride or in water. A
known
112
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
amount of one or more active agents to be incorporated in the particles is
suspended
(in the case of an insoluble active agent) or co-dissolved (in the case of a
soluble
active agent) in the polymer solution. The solution or dispersion is pumped
through a
micronizing nozzle driven by a flow of compressed gas, and the resulting
aerosol is
suspended in a heated cyclone of air, allowing the solvent to evaporate from
the
microdroplets, forming particles. Microspheres/nanospheres ranging between 0.1-
10
microns can be obtained using this method.
c. Interfacial Polymerization
Interfacial polymerization can also be used to encapsulate one or more active
agents.
Using this method, a monomer and the active agent(s) are dissolved in a
solvent. A
second monomer is dissolved in a second solvent (typically aqueous) which is
immiscible with the first. An emulsion is formed by suspending the first
solution through
stirring in the second solution. Once the emulsion is stabilized, an initiator
is added to
the aqueous phase causing interfacial polymerization at the interface of each
droplet
of emulsion.
d. Hot Melt Microencapsulation
Microspheres can be formed from polymers such as polyesters and polyanhydrides
using hot melt microencapsulation methods as described in Mathiowitz etal.,
Reactive
Polymers, 6:275 (1987). In this method, the use of polymers with molecular
weights
between 3-75,000 daltons is preferred. In this method, the polymer first is
melted and
then mixed with the solid particles of one or more active agents to be
incorporated that
have been sieved to less than 50 microns. The mixture is suspended in a non-
miscible
solvent (like silicon oil), and, with continuous stirring, heated to 5°
C. above the
melting point of the polymer. Once the emulsion is stabilized, it is cooled
until the
polymer particles solidify. The resulting microspheres are washed by decanting
with
petroleum ether to give a free-flowing powder.
e. Phase Separation Microencapsulation
In phase separation microencapsulation techniques, a polymer solution is
stirred,
optionally in the presence of one or more active agents to be encapsulated.
While
continuing to uniformly suspend the material through stirring, a nonsolvent
for the
polymer is slowly added to the solution to decrease the polymer's solubility.
Depending
on the solubility of the polymer in the solvent and nonsolvent, the polymer
either
113
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
precipitates or phase separates into a polymer rich and a polymer poor phase.
Under
proper conditions, the polymer in the polymer rich phase will migrate to the
interface
with the continuous phase, encapsulating the active agent(s) in a droplet with
an outer
polymer shell.
f. Spontaneous Emulsion Microencapsulation
Spontaneous emulsification involves solidifying emulsified liquid polymer
droplets
formed above by changing temperature, evaporating solvent, or adding chemical
cross-linking agents. The physical and chemical properties of the encapsulant,
as well
as the properties of the one or more active agents optionally incorporated
into the
nascent particles, dictates suitable methods of encapsulation. Factors such as
hydrophobicity, molecular weight, chemical stability, and thermal stability
affect
encapsulation.
g. Solvent Evaporation Microencapsulation
Methods for forming microspheres using solvent evaporation techniques are
described in E. Mathiowitz et at,Scanning Microscopy, 4:329(1990); L. R. Beck
et at,
Fertil. Steril., 31:545 (1979); L. R. Beck et al Am J Obstet Gynecol 135(3)
(1979); S.
Benita et at ,Pharm. Sci., 73:1721 (1984); and U.S. Pat. No. 3,960,757 to
Morishita et
al. The polymer is dissolved in a volatile organic solvent, such as methylene
chloride.
One or more active agents to be incorporated are optionally added to the
solution, and
the mixture is suspended in an aqueous solution that contains a surface active
agent
such as poly(vinyl alcohol). The resulting emulsion is stirred until most of
the organic
solvent evaporated, leaving solid microspheres/nanospheres. This method is
useful
for relatively stable polymers like polyesters and polystyrene. However,
labile
polymers, such as polyanhydrides, may degrade during the fabrication process
due to
the presence of water. For these polymers, some of the following methods
performed
in completely anhydrous organic solvents are more useful.
h. Solvent Removal Microencapsulation
The solvent removal microencapsulation technique is primarily designed for
polyanhydrides and is described, for example, in WO 93/21906 to Brown
University
Research Foundation. In this method, the substance to be incorporated is
dispersed
or dissolved in a solution of the selected polymer in a volatile organic
solvent, such as
methylene chloride. This mixture is suspended by stirring in an organic oil,
such as
114
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
silicon oil, to form an emulsion. Microspheres that range between 1-300
microns can
be obtained by this procedure. Substances which can be incorporated in the
microspheres include pharmaceuticals, pesticides, nutrients, imaging agents,
and
metal compounds.
i. Coacervation
Encapsulation procedures for various substances using coacervation techniques
are
known in the art, for example, in GB-B-929 406; GB-B-929 40 1; and U.S. Pat.
Nos.
3,266,987, 4,794,000, and 4,460,563. Coacervation involves the separation of a
macromolecular solution into two immiscible liquid phases. One phase is a
dense
coacervate phase, which contains a high concentration of the polymer
encapsulant
(and optionally one or more active agents), while the second phase contains a
low
concentration of the polymer. Within the dense coacervate phase, the polymer
encapsulant forms nanoscale or microscale droplets. Coacervation may be
induced
by a temperature change, addition of a non-solvent or addition of a micro-salt
(simple
coacervation), or by the addition of another polymer thereby forming an
interpolymer
complex (complex coacervation).
j. Low Temperature Casting of Microspheres
Methods for very low temperature casting of controlled release microspheres
are
described in U.S. Pat. No. 5,019,400 to Gonnbotz et al. In this method, a
polymer is
dissolved in a solvent optionally with one or more dissolved or dispersed
active agents.
The mixture is then atomized into a vessel containing a liquid non-solvent at
a
temperature below the freezing point of the polymer-substance solution which
freezes
the polymer droplets. As the droplets and non-solvent for the polymer are
warmed, the
solvent in the droplets thaws and is extracted into the non-solvent, resulting
in the
hardening of the microspheres.
k. Phase Inversion Nanoencapsulation (PIN)
Nanoparticles can also be formed using the phase inversion nanoencapsulation
(PIN)
method, wherein a polymer is dissolved in a "good" solvent, fine particles of
a
substance to be incorporated, such as a drug, are mixed or dissolved in the
polymer
solution, and the mixture is poured into a strong non-solvent for the polymer,
to
spontaneously produce, under favorable conditions, polymeric microspheres,
wherein
the polymer is either coated with the particles or the particles are dispersed
in the
115
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
polymer. See, e.g., U.S. Pat. No. 6,143,211 to Mathiowitz, et at The method
can be
used to produce monodisperse populations of nanoparticles and microparticles
in a
wide range of sizes, including, for example, about 100 nanometers to about 10
microns.
Advantageously, an emulsion need not be formed prior to precipitation. The
process
can be used to form microspheres from thermoplastic polymers.
I. Sequential Phase Inversion Nanoencapsulation (sPIN)
Multi-walled nanoparticles can also be formed by a process referred to herein
as
"sequential phase inversion nanoencapsulation" (sPIN). This process is
described in
detail below in Section IV. sPIN is particularly suited for forming
monodisperse
populations of nanoparticles, avoiding the need for an additional separations
step to
achieve a monodisperse population of nanoparticles.
m. Dissolving Tablet
In certain embodiments, the anti-PESC agents is provided in a dissolving
tablet. For
example, the tablet can contain a therapeutically effective amount of the anti-
PESC
agent in combination with polyvinylpyrrolidone (PVP: povidone), wherein the
tablet is
formulated to rapidly dissolve in a specific volume of liquid so as to
generate a topical
esophageal therapy suitable for delivering the anti-PESC to the luminal
surface of the
esophagus.
For instance, the the volume of liquid in which the tablet dissolves can be
from 5 to 50
mL, 5 to 25 mL or even 5 to 15 mL. Preferably the liquid is water.
The dissolving tablet can also further include an excipient that renders the
dissolving
tablet palatable, especially at least one excipient that increases viscosity
of the topical
esophageal therapy. An exemplary viscosity-enhancing excipient is mannitol.
n. Topical Formulation
In certain embodiments, the anti-PESC agent is provided in a topical, non-
systemic,
oral, slow releasing, solid, soft lozenge pharmaceutical composition
comprising: (a)
about 1% to about 5% by mass of one or more release modifiers comprising
polyethylene oxide polymers comprising a molecular weight of about 900,000 to
about
8,000,000; (b) about 10% to about 60% by mass of one or more film-forming
polymers
comprising gelatins; (c) about 5% to about 20% by mass of one or more
plasticizers
116
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
comprising glycerol, sorbitol, or combinations thereof; and (d) less than 1%
by mass
of one or more anti-PESC agents. Exemplary plasticizers include glycerol,
sorbitol,
mannitol, maltitol, xylitol, or combinations thereof. The lozenge may also
include one
or more sweeteners, such as maltitol, xylitol, mannitol, sucralose, aspartame,
stevia,
or a combination thereof. The lozenge may also include one or more pH
modifiers
comprising one or more organic acids.
VII. Examples
Barrett's Esophagus (BE) is an irreversible condition that is believed to be
the
precancerous lesion of Esophageal Adenocarcinoma (EAC). BE originates from a
unique cell population preexisting at gastroesophageal junction and possesses
its own
stem cells. Using a novel ground-state stem cell cloning technology, we
derived
patient-matched stem cell pedigree lines from BE and Esophageal epithelia.
Image-
based high-throughput chemical screening was developed and used to uncover a
unique combination of chemicals that can specifically eradicate BE stem cells
while
protecting and promoting Esophageal stem cells. These effects were confirmed
in a
co-culture 3D model and a mouse xenograft model by co-transplanting BE and
Esophageal stem cells in vivo. Interestingly, this drug combination was also
able to
eradicate stem cells in patient-matched dysplasia and cancer. It is the first
time that
ground-state stem cells from patient-matched BE, dysplasia, cancer and normal
tissue
can be cloned and expanded in vitro and employed to identify targeted
therapeutics
for not only preemptive therapies of BE but therapies targeting late-stage
cancers. It
is anticipated that this platform can be applied to provide the promise of
developing
novel strategies for chemoprevention and treatment of various types of lethal
cancers.
117
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Cloning Barrett's Esophagus Stem Cells and Chemical Screening
Endoscopic mucosal biopsies obtained from the distal esophagus of Barrett's
patients
can include Esophageal squamous epithelium and Barrett's Esophagus (Fig. 1A;
Yamamoto et al). Colonies arose from single cell suspensions of these 1mm
biopsies
one week after plating onto lawns of irradiated 3T3 cells in growth media
known to
support immature, epithelial stem cells (Wang et at, 2015; Yamamoto et at,
2016;
Duleba etal., 2018) (Fig. 1A; StemECHO, MCT). As reported previously (Yamamoto
et at, 2016), the colonies yielded from Barrett's yielded mixtures of both
Krt5-positive
clones typical of the esophageal squamous basal cells and ones that expressed
the
columnar epithelial marker Krt7 (Fig. 1A). To separate these two populations
of clones
derived from the Barrett's biopsies, multiple single colonies were samples and
expanded as independent pedigrees (Yamamoto et al., 2016) (Fig. 1A). Reprobing
these pedigree lines with the same antibodies showed that the original
Barrett's
biopsies contained two distinct clonogenic cells marked by committed
expression of
either Krt5 or Krt7 (Fig.1A).
In order to explore the possibility of uncovering an agent that can
selectively
eradicate Barrett's stem cells, a high throughput chemical screening on a
panel of
esophageal squamous stem cells and Barrett's stem cells was set up. Three
libraries
were used for this screening including Custom Clinical, Prestwick and Selleck
in totally
of non-overlapping 2276 chemicals (Fig. 1B). While approximately 70% of the
chemicals had no impact on either of the cell lines and around 10% of the
chemicals
eradicate both cell types, the other 20% of the chemicals displayed
differential efforts
on these two cell types (Figs. 1C, IF and 1G). The selected hits were
validated in a
dosage-response manner, suggesting the reliability of the screening approach
(Figs.
1F and 1G). Among these 20% of the chemicals, we did not observe one single
agent
that can play dual roles including eradication of Barrett's stem cells and
protection of
esophageal stem cells. Interestingly, Ponatinib was identified to
significantly increase
the proliferation of esophageal stem cells while slightly decreasing the
survival of
Barrett's stem cells (Fig. 1D and E).
Synthetic Chemical Screening for Dual Action Regime
118
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
It was next hypothesized that one chemical in our libraries could function
with
Ponatinib in a synergistic manner to specifically eradicate Barrett's stem
cells while
protecting and promoting esophageal stem cells. The same libraries were
screened
in the background of 1pM Ponatinib. Interestingly, eight compounds displayed
the
lethality towards Barrett's stem cells without affecting Esophageal stem cells
in the
presence of Ponatinib (Fig. 2A). Among them, Adefovir Dipivoxil is a FDA
approved
drug, CEP-18770 and Qizartinib are in clinical trials. JIB04, VVZ8040,
Stattic,
Nanchangmycin and AZD1080 are not in any clinical trials (Fig. 2B). None of
these
chemicals have been reported to target BE and interestingly, they are
targeting various
different pathways and harbor distinct chemical structures. Their differential
effects
on Barrett's stem cells and Esophageal stem cells were validated in a dosage
dependent manner (Fig_ 2C; Fig_ 3F). Moreover, co-culture of nine Barrett's
stem cell
lines from patients with spectrum of mutation profiles (Yamamoto et al, 2016)
and
Esophageal stem cells demonstrated CEP-18770, JIB04, in the presence of
Ponatinib
displayed uniform lethality to all Barrett's stem cells that we tested while
promoting
Esophageal squamous stem cells (Fig. 2D). This result suggests that these two
chemicals could work synergistically with Ponatinib to target Barrett's stem
cells
specifically independent of the genonnics and stage of these Barrett's stem
cells. The
combination of Nanchangmycin and Ponatinib was not pursued due to the high
lethality of 3T3-J2 feeder cells during the treatment, which suggests
potential toxicity
towards other types of somatic cells in viva
Validating Dual Action Regime in 3D culture and Mouse Xenograft Model
A 3D culture model was generated to mimic Barrett's Esophagus in vitro. The
esophageal squamous stem cells and Barrett's stem cells were co-cultured in
the
transwell insert. Following the creation of air-liquid interface, it was
observed that the
BE islands located among well-differentiated squamous epithelium that well
recapitulated the histology of human BE in the patient. Given the similarity
of this
artificial model and human pathology, the hits were tested in this system to
further
validate them. Eight Barrett's stem cell lines were co-cultured with
Esophageal stem
cells in ALI system and CEP-18770 or JIB04 together with Ponatinib were added
in
the medium following the creation of ALI. The esophageal squamous stem cells
were
labeled with red fluorescent protein (RFP) while Barrett's stem cells were
labeled with
119
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
green fluorescent protein (GFP). Without the treatment, it was observed that a
mixture
of GFP and RFP labeled cells in culture. However, following the treatment of
either
CEP-18770 or JIB04 in the presence of Ponatinib, the disappearance of GFP
labeled
Barrett's stem cells was observed. The cross sections of the ALI structures
showed
intact esophageal squamous epithelium without any existence of Barrett's
epithelium
(Fig. 2 A&B).
The effect of these chemicals on BE in a xenograft model was further examined.
The
esophageal squamous stem cells and GFP labeled Barrett's stem cells were mixed
and co-injected into the immunodeficient mice. After five days of xenograft,
the
Ponatinib and CEP-18770 combination or Ponatinib and JIB04 combination was
injected into the mice intraperitoneally in an alternative manner (Fig. 3C).
Two weeks
following the treatment, the xenografts were collected and examined through
histology
analysis and clonogenic analysis. It was found that the total loss of
Barrett's structures
in the treated animals, which is consistent with the lack of clonogenic
Barrett's stem
cells in vitro (Fig. 3D and 3E; Fig. 4E; Figs. 5F & 5G).
Stem Cells of Barrett's Esophagus, Dysplasia and Cancer Can Be Targeted
Similarly
BE has been known to be the precursor of the dysplastic and cancerous lesions
of
EAC. The transcriptonrie analysis of patient-matched Barrett's, dysplastic and
EAC
stem cells revealed significantly overlapping genes and pathways in comparison
to the
stem cells derived from Esophagus squamous epithelium. Moreover, PCA analysis
of
Barrett's stem cells derived from 12 patients together with dysplasia, cancer
and
Esophageal squamous stem cells confirmed that the stem cells of dysplasia and
cancer shared very similar gene expression with Barrett's stem cells (Fig. 4 A
& B).
This data suggests that the strategies developed to eradicate BE could also be
used
to target dysplasia and EAC which have routinely relapsed from the traditional
chemotherapies. The effects of the eight drug hits in combination with
Ponatinib were
tested in the cell culture of co-seeding esophageal stem cells and Barrett's,
dysplasia
or EAC stem cells. Most of these combinations could eliminate all three
entities
without affecting esophageal squamous stem cells in two independent patients
(Fig.
4C). Importantly, the JIB04 and CEP-18770 can function in a synergistic manner
with
120
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Ponatinib to eradicate precancerous, dysplastic and cancerous stem cells of
EAC in
both 3D culture system and mouse xenograft models (Fig. 5). Given these three
entities always co-exist in patients with EAC, our strategies of eradicating
all of them
could help to eliminate the chance of recurrence of EAC patients drastically.
Discussion of Results
Despite its influential position in cancer prevention, and fundamental
advances in
ablative approaches aiming to eradicate it preemptively, Barrett's remains an
enormous and growing problem with an estimated 3 million cases in the US
alone.
Advanced BE lesions that are treated by ablative therapies recur at alarming
rates
(reference), suggesting an unmet medical need for effective and targeted
therapeutic
options for these patients. The ability to clone and expand the ground-state
stem cells
of BE, dysplasia, EAC and normal esophageal squamous epithelium from matched
patient samples provides a very promising platform for high-throughput
chemical
screening devoted to developing highly selective means of eradicating BE and
more
advanced lesions ahead of the onset of cancer. Each of these stem cell clones
can
be individually differentiated by polarization in so-called "air-liquid
interface" cultures
to yield 3-D epithelia remarkably similar to that of the in situ normal,
lesional or
cancerous epithelia. These same stem cell clones were used in advanced co-
culture
models with normal epithelial stem cells to investigate the potential ability
of selected
drug combinations to alter the competitive status of such lesions in the
distal
esophagus. Furthermore, a novel mouse model for testing drug combinations in
vivo
was established by transplanting patient-derived BE, dysplasia, EAC and
esophageal
squamous stem cells in the NSG mice subcutaneously.
The first direction of chemical screening focused on single agents that showed
a
greater effect against BE stem cells than the patient-matched esophageal stem
cells
as that was a key goal of this effort_ However, upon detailed dose-response
studies of
these single agent "hits", the standard differential at the optimal dosing
was, at best,
10-fold in concentration. This 10-fold differential in concentration between
lethal
effects on BE stem cells and those of normal esophagus was disappointing in
that it
suggested a relatively narrow therapeutic window.
The potential of the small molecules that selectively favored the growth of
the normal
esophageal stem cells was explored. The intestinal metaplasia of BE is thought
to be
121
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
in competition with the surrounding esophageal mucosa during its proximal
spread
(Wang et at, 2011). Thus, in any therapeutic strategy, it is possible that
this
competitive interaction could be exploited from the standpoint of improving
the
competitive edge of the esophagus. Therefore, it is hypothesized that the
molecules
found to promote the growth of esophageal stem cells, such as Ponatinib could
be
used in combination with molecules that limit BE stem cells. Established
herein are
"synthetic lethal" screens (McCormick, 2015; Thompson et at, 2017; Aguirre and
Hahn, 2018) against BE stem cells using small molecule libraries in a
background of
Ponatinib. Interestingly, this synthetic lethal screen identified a different
set of small
molecules that, in the context of Ponatinib, efficiently kill BE stem cells
while at the
same time augmented the growth of normal esophageal stem cells. Among them,
..11B04 and CEP-18770 in a synergistic manner with Ponatinib were most
effective in
eradicating BE stem cells of a wide-range of patients. Most important from the
standpoint of potential therapeutics, dose-response curves for these anti-BE
stem cell
components of this combination, in the background of a fixed concentration of
the
Ponatinib, shows a remarkable differential of nearly 1,000-fold.
Significant progress has been made in mimicking the competitive interactions
between
BE and esophageal stem cells through the generation of co-cultures in vitro.
Thus,
"squamous islands" (Sharma et at, 1998) of esophageal epithelia interspersed
with
regions dominated by Barrett's esophagus can be generated. These steady state
co-
cultures have been used to test the impact of the synthetic lethal drug
combinations
identified in small molecule screens. Significantly, these drug combinations
have
proven to be remarkably effective in eliminating the Barrett's esophagus and
permitting
the esophageal stem cells to expand to fill in the regions vacated by the loss
of the BE
epithelia. Similar efforts were also made to model the competitive
interactions between
Barrett's and esophageal epithelia in vivo in xenografts. Remarkably, these
two drug
combinations showed effects on dysplasia and EAC stem cells with similar
selectivity
and doses as those towards Barrett's stem cells. The large differential
between doses
of these two effective combinations that eliminated BE, dysplasia, and EAC
stem cells
versus normal esophageal stem cells suggest the possibility that such drug
combinations could produce larger "therapeutic windows" than we observed for
single
agents directed at the Barrett's alone_
122
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
Therefore, in addition to identifying synthetic lethal drug combinations
effective for BE,
these compounds are shown to also be effective for targeting progenitor cells
of
dysplasia and EAC that were not part of the initial screen. This finding
raises the
question as to whether these compounds are targeting some feature of the
Correa
sequence "lineage" rather than traditional chemotherapeutics that target DNA
synthesis or common activities of a rapidly dividing cell.
Taken together, precancerous and cancerous lesions are regenerative and like
normal
regenerative epithelia depend on discrete populations of immature stem cells
for this
regenerative growth, and that therapeutics directed at these stem cell
populations
could be highly specific and effective approaches to eliminating these lesions
for
therapeutic benefit. Therefore, this platform and similar approaches can be
applied to
drug discovery efforts in other cancer types.
Methods
High throughput screening and imaging of selected cell lines
The GFP-tagged cell lines will be seeded on multiple 384 well plates (Griener
Bio-
One, USA) (varying according to number of compounds in a library) with a
feeder layer
of irradiated 3T3-J2 fibroblast cells. The stem cell will be allowed to grow
until they
divide to become 4-5 cells within a colony. Afterwards they will be
transported for
treatment with selected chemical library (1 pM) to High Throughput Research
and
Screening Center at Institute of Biosciences and Technology (Houston, Texas),
Texas
A&M University. Positive and negative control lanes will be allocated within
each plate.
A highly potent drug will be used as a positive control and negative control
will be just
with DMSO since most of the drugs are dissolved in DMSO. The cells will be
allowed
to grow for 6 days at 37 C, 7.5% CO2 incubator after treatment. After the
cells in the
control lanes are at good confluency, the cells will be prepared for imaging.
Each multi-
well plates will be washed with Phosphate Buffered Saline (Gibco, USA) and
fixed with
4% paraformaldehyde at room temperature for 25 minutes. Paraformaldehyde will
be
replaced with Phosphate Buffered Saline (PBS) and will be imaged using
Inverted
Eclipse Ti-Series (Nikon, Japan) microscope with Lumencor SOLA light engine,
paired
with High Content Microscope system (Nikon, Japan), Andor Technology Clara
Interline CCD camera, NIS-Elements Advanced Research v.4.13 software (Nikon,
Japan) and NIS-Elements HC software (Nikon, Japan). High Content Analysis
(HCA)
123
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
system built on NIS-Elements platform streamlined with automated well plate
acquisition and multiple well plate job run will be used for high throughput
imaging of
phase contrast as well as FITC channel.
High Content Image Analysis, selection of drugs, and dose-response analysis
NIS-Elements High Content Analysis (HCA) system will be used for image data
management of multiple well plate job runs. The cells labelled with Green
Fluorescent
Protein (GFP) in each well of multi well plates will be imaged with features
of each
stem cell colonies. The changes in cell phenotype compared to control lanes
will be
measured based on the fluorescent signal threshold using automated image
analysis.
The features like area, colony number will be exported from the automated
analysis.
Treated wells will be compared to untreated wells based on GFP-signal area and
number of stem cells colonies. The treated well will be normalized with the
untreated
well based on the area which will be represented in terms of survival rate
(percentage
of contrail) and will be compared between control and pathogenic population of
cells.
Z'-factor2,3 will be calculated based on the difference between positive and
negative
control and will be used as criteria for assessing quality of runs. Only
plates with 1-
factor >0.6 will be used. The compounds with a coefficient of variation
between plate
duplicates larger than 20% will be ignored. B-scorel will be calculated via R
package
platetools v0Ø2 (github.com/swarchaliplatetools) to control the edge and
positional
bias to help infer the potential hits of inhibitors. Only the compounds with a
cutoff of B-
score < -2 will be considered as potential inhibitors. The selection of drugs
will be
made based on the maximum differences in survival rate between patient-matched
cell lines (cut-off set at 20%), their targets, structural spectrum, pathways
related to
them and their possible relation implicated in Barret's esophageal. If there
are not
patient matched pedigrees, the median value of survival rates in a contain
group will
be used as representative value for that group of pedigrees. The dose-response
curves of survival rate for a certain compound will be calculated by fitting a
three-
parameter log-logistic dose-response model to the survival rate data using R
package
dre v3Ø1. R packages !attests v0.9-36 and sandwich6 v2.4-0 to obtain robust
standard errors to address the fact that some variance heterogeneity is
present. The
ED50 value is estimated by module ED in the R package drc.
124
CA 03135020 2021- 10-25
WO 2020/219963
PCT/US2020/029933
VIII. References
1. Malo, N., Hanley, J.A., Cerquozzi, S., Pelletier, J. & Nadon, R.
Statistical
practice in high-throughput screening data analysis. Nat Biotechnol 24, 167-
75(2006).
2. Zhang, J.H., Chung, T.D. & Oldenburg, K.R. A Simple Statistical
Parameter for
Use in Evaluation and Validation of High Throughput Screening Assays. J Blomol
Screen 4, 67-73 (1999).
3. Iversen, P.W., Eastwood, B.J., Sittampalam, G.S. & Cox, K.L. A
comparison of
assay performance measures in screening assays: signal window, Z factor, and
assay
variability ratio. J Biomol Screen 11, 247-52 (2006).
4. Ritz, C., Baty, F., Streibig, J.C. & Gerhard, D. Dose-Response
Analysis Using
R. Plos One 10(2015).
5. Zeileis, A. & Hothom, T. Diagnostic Checking in Regression
Relationships. R
News 2, 7-10 (2002).
6. Zeileis, A. Econometric Computing with HC and HAC Covariance Matrix
Estimators. Journal of Statistical Software 1, 1-17 (2004).
125
CA 03135020 2021- 10-25